63 results on '"Amanda Saratsis"'
Search Results
2. Data from Methylation-dependent Tissue Factor Suppression Contributes to the Reduced Malignancy of IDH1-mutant Gliomas
- Author
-
Craig Horbinski, C. David James, Jann N. Sarkaria, Denise M. Scholtens, Amanda Saratsis, Kathleen McCortney, Rodrigo Javier, Qazi F. Haider, Yuping D. Li, Jonathan Lamano, Michael Drumm, Brian Wray, Snezana Mirkov, and Dusten Unruh
- Abstract
Purpose:Gliomas with isocitrate dehydrogenase 1 mutations (IDH1mut) are less aggressive than IDH1 wild-type (IDH1wt) gliomas and have global genomic hypermethylation. Yet it is unclear how specific hypermethylation events contribute to the IDH1mut phenotype. Previously, we showed that the gene encoding the procoagulant tissue factor (TF), F3, is among the most hypermethylated and downregulated genes in IDH1mut gliomas, correlating with greatly reduced thrombosis in patients with IDH1mut glioma. Because TF also increases the aggressiveness of many cancers, the current study explored the contribution of TF suppression to the reduced malignancy of IDH1mut gliomas.Experimental Design: TF expression was manipulated in patient-derived IDH1mut and IDH1wt glioma cells, followed by evaluation of in vitro and in vivo behavior and analyses of cell signaling pathways.Results:A demethylating agent, decitabine, increased F3 transcription and TF-dependent coagulative activity in IDH1mut cells, but not in IDH1wt cells. TF induction enhanced the proliferation, invasion, and colony formation of IDH1mut cells, and increased the intracranial engraftment of IDH1mut GBM164 from 0% to 100% (P = 0.0001). Conversely, TF knockdown doubled the median survival of mice engrafted with IDH1wt/EGFRvIIIamp GBM6, and caused complete regression of IDH1wt/EGFRamp GBM12 (P = 0.001). In vitro and in vivo effects were linked to activation of receptor tyrosine kinases (RTK) by TF through a Src-dependent intracellular pathway, even when extracellular RTK stimulation was blocked. TF stimulated invasion predominately through upregulation of β-catenin.Conclusions:These data show that TF suppression is a component of IDH1mut glioma behavior, and that it may therefore be an attractive target against IDH1wt gliomas.
- Published
- 2023
3. Supplementary Data Figures from Methylation-dependent Tissue Factor Suppression Contributes to the Reduced Malignancy of IDH1-mutant Gliomas
- Author
-
Craig Horbinski, C. David James, Jann N. Sarkaria, Denise M. Scholtens, Amanda Saratsis, Kathleen McCortney, Rodrigo Javier, Qazi F. Haider, Yuping D. Li, Jonathan Lamano, Michael Drumm, Brian Wray, Snezana Mirkov, and Dusten Unruh
- Abstract
Supplementary data figures
- Published
- 2023
4. Combinatorial Inhibition of Epigenetic Regulators to Treat Glioblastoma
- Author
-
Noah Burket, Jenna Koenig, and Amanda Saratsis
- Subjects
Ocean Engineering - Abstract
Background: Glioblastoma multiforme (GBM) is a deadly primary brain cancer that is diagnosed in 12,000 patients in the US annually with a median survival time of 15 months. Temozolomide is the standard-of-care for GBM; however, many tumors are resistant, necessitating the expansion of therapeutic options. EZH2 and JMJD3 are two proteins responsible for epigenetic regulation of the genome via histone methylation, with EZH2 also affecting non-histone targets. Prior studies showed that inhibition of these proteins decreased cell counts and induced radiosensitivity in GBM. Thus, we investigated combined use of EZH2 inhibitor, EPZ-6438, and JMJD3 inhibitor, GSK-J4, in the treatment of temozolomide-resistant GBM10 cells. Methods: Non-irradiated cells were treated with both drugs singly or combined, and counted at 24-, 48-, and 72-hour intervals. Irradiated cells were pre-treated with each drug or combination therapy for three days, irradiated, and then counted at 24-, 48-, and 72-hour intervals. Western blot allowed investigation of dsDNA damage biomarker yH2AX, gene-silencing modification H3K27me3, total H3, tumor suppressor p53, EZH2, JMJD3, ySTAT3, and total STAT3 expression in non-irradiated and irradiated cells following drug treatment. Results: Single EPZ-6438 and GSK-J4 treatment decreased cell count in a dose and time dependent manner. GSK-J4 was more effective than EPZ-6438, and combinatorial treatment was most effective. Western blot revealed that GSK-J4 but not EPZ-6438 treatment followed by radiation increased H3K27me3 expression. EPZ-6438 treatment increased yH2AX expression, but this was not further increased by radiation. Meanwhile, GSK-J4 treatment increased yH2AX, but only after radiation. Discussion: Decreased cell count following GSK-J4 treatment may be due to increased gene silencing resulting from the inhibition of H3K27 demethylation. Additionally, increased dsDNA breaks observed in EPZ-6438 and GSK-J4 treatments supports their roles in radiosensitizing GBM cells. Potential Impact: This study highlights the importance of further investigation into GSK-J4 and EPZ-6438 combination therapy in temozolomide-resistant GBM tumors.
- Published
- 2023
5. Gorham Stout disease of the temporal bone with cerebrospinal fluid leak
- Author
-
Amanda Saratsis, Pascale Aouad, Maura E. Ryan, Nancy M. Young, and Meredith A Reynolds
- Subjects
Pathology ,medicine.medical_specialty ,Osteolysis ,Cerebrospinal fluid leak ,business.industry ,General Medicine ,medicine.disease ,Resorption ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Langerhans cell histiocytosis ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Lymphangioma ,Temporal bone ,medicine ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Rare disease - Abstract
Gorham Stout disease (GSD) is a rare disease characterized by the proliferation of endothelial lined vessels and replacement of bone by fibrous tissue. The main imaging features are progressive osteolysis and cortical resorption. Temporal bone involvement is rare but presents as a destructive bone lesion that may be misinterpreted as more common lytic processes in the pediatric population, such as infection or Langerhans cell histiocytosis. GSD of the temporal bone is associated with cerebrospinal fluid (CSF) leaks, may present with otorrhea, and can mimic other causes of ear drainage. Here, we report the clinical course, imaging features, and outcomes of a 3-year-old girl with GSD of the temporal bone presenting with CSF leak initially attributed to infection.
- Published
- 2021
6. Safety of Ventricular Reservoir Sampling in Pediatric Posthemorrhagic Hydrocephalus Patients: Institutional Experience and Review of the Literature
- Author
-
Amanda Saratsis, Tord D. Alden, Arthur J DiPatri, Tadanori Tomita, Maureen Kilgallon, Stacy Speck, Robin Bowman, Daphne Li, Sandi Lam, and Kathy Romanski
- Subjects
medicine.medical_specialty ,MEDLINE ,Ventricular reservoir ,030204 cardiovascular system & hematology ,Ventriculoperitoneal Shunt ,Asepsis ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Posthemorrhagic hydrocephalus ,Risk Factors ,medicine ,Humans ,Sampling (medicine) ,Child ,Cerebral Hemorrhage ,Retrospective Studies ,Endocrine and Autonomic Systems ,business.industry ,Incidence (epidemiology) ,Infant, Newborn ,Infant ,Surgery ,Medical–Surgical Nursing ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Hydrocephalus ,Medical literature - Abstract
INTRODUCTION: Posthemorrhagic hydrocephalus (PHH) is a common disease process encountered in neonates. Management often includes cerebrospinal fluid (CSF) aspiration through ventricular access devices (VADs). However, a common concern surrounding serial access of implanted subcutaneous reservoirs includes introduction of infection. In addition, there is great variability in aseptic technique. Therefore, the authors sought to evaluate the incidence of VAD access-associated infections in the literature and compare them with the rate of infection found at our institution. We also highlight the use of a preassembled VAD access kit and standardized access protocol, as well as the role of provider education, in maintaining safety and sterility during serial VAD access. METHODS: A single-institution retrospective review was performed for PHH patients younger than 1 year old undergoing serial CSF aspirations via implanted VADs (2009-2019). Patients were excluded if they had a ventriculoperitoneal shunt placed as primary intervention. MEDLINE search for reports of serial VAD access in PHH was also performed. Reports were excluded if they did not include full-text articles in the English literature. RESULTS: At our institution, subcutaneous reservoirs were placed in 37 neonates with PHH for serial CSF aspiration. No infections occurred after a total of 630 taps (average, 17 taps per reservoir; range, 0-83) and 10 420 collective reservoir days (average, 282 per patient; range, 6-3700). Only 2 reservoirs required revision for malfunction. Serial VAD taps for PHH were described in 14 articles in the medical literature, with 7.9% (n = 47/592) of patients reported with tap-related infectious complications. CONCLUSION: A standardized VAD access kit, along with stringent adherence to access protocol, can significantly minimize risk of infection associated with serial VAD access. These principles can be generalized to percutaneous aspiration of CSF from subcutaneous reservoirs placed for other indications to promote safety and sterility of this common procedure.
- Published
- 2021
7. Therapeutic targeting of transcriptional elongation in diffuse intrinsic pontine glioma
- Author
-
Amanda Saratsis, Edwin R. Smith, Yongzhan Zhang, Gavin T. Blyth, Hiroaki Katagi, Nozomu Takata, Akihide Kondo, Stewart Goldman, Lihua Zou, Yuki Aoi, Rintaro Hashizume, Yusuke Tomita, Frank Eckerdt, Ali Shilatifard, Oren J. Becher, Takahiro Sasaki, and Emily J. Rendleman
- Subjects
Cancer Research ,biology ,Cell growth ,Chemistry ,RNA polymerase II ,Cell cycle ,Chromatin ,Oncology ,Apoptosis ,Transcription (biology) ,Gene expression ,Cancer research ,biology.protein ,Neurology (clinical) ,Viability assay - Abstract
BackgroundDiffuse intrinsic pontine glioma (DIPG) is associated with transcriptional dysregulation driven by H3K27 mutation. The super elongation complex (SEC) is required for transcriptional elongation through release of RNA polymerase II (Pol II). Inhibition of transcription elongation by SEC disruption can be an effective therapeutic strategy of H3K27M-mutant DIPG. Here, we tested the effect of pharmacological disruption of the SEC in H3K27M-mutant DIPG to advance understanding of the molecular mechanism and as a new therapeutic strategy for DIPG.MethodsShort hairpin RNAs (shRNAs) were used to suppress the expression of AF4/FMR2 4 (AFF4), a central SEC component, in H3K27M-mutant DIPG cells. A peptidomimetic lead compound KL-1 was used to disrupt a functional component of SEC. Cell viability assay, colony formation assay, and apoptosis assay were utilized to analyze the effects of KL-1 treatment. RNA- and ChIP-sequencing were used to determine the effects of KL-1 on gene expression and chromatin occupancy. We treated mice bearing H3K27M-mutant DIPG patient-derived xenografts (PDXs) with KL-1. Intracranial tumor growth was monitored by bioluminescence image and therapeutic response was evaluated by animal survival.ResultsDepletion of AFF4 significantly reduced the cell growth of H3K27M-mutant DIPG. KL-1 increased genome-wide Pol II occupancy and suppressed transcription involving multiple cellular processes that promote cell proliferation and differentiation of DIPG. KL-1 treatment suppressed DIPG cell growth, increased apoptosis, and prolonged animal survival with H3K27M-mutant DIPG PDXs.ConclusionsSEC disruption by KL-1 increased therapeutic benefit in vitro and in vivo, supporting a potential therapeutic activity of KL-1 in H3K27M-mutant DIPG.
- Published
- 2021
8. Patient and operative factors associated with unanticipated intensive care admission and outcomes following posterior fossa decompressions in children: A retrospective study
- Author
-
Hubert A. Benzon, Anthony Tantoco, Anthony Longhini, John Hajduk, Amanda Saratsis, Santhanam Suresh, Robert J. McCarthy, and Narasimhan Jagannathan
- Subjects
Decompression ,Anesthesiology and Pain Medicine ,Postoperative Complications ,Treatment Outcome ,Critical Care ,Pediatrics, Perinatology and Child Health ,Humans ,Child ,Intraoperative Complications ,Arnold-Chiari Malformation ,Retrospective Studies - Abstract
Posterior fossa decompression for Chiari I Malformation is a common pediatric neurosurgical procedure. We sought to identify the impact of anesthesia-related intraoperative complications on unanticipated admission to the intensive care unit and outcomes following posterior fossa decompression.Medical records of all patients18 years who underwent surgery for Chiari I malformation between 1/1/09 and 1/31/21 at the AnnRobert H. Lurie Children's Hospital of Chicago were included. Records were reviewed for patient characteristics, anesthesia-related intraoperative complications, postoperative complications, and surgical outcomes. The primary outcome was the incidence of unanticipated admission to the intensive care unit, and the primary variable of interest was an anesthesia-related intraoperative complication. Patient, surgical characteristics, and year of surgery were also compared between patients with and without an unanticipated admission to the intensive care unit, and a multi-variable adjusted estimate of odds of unanticipated admission to the intensive care unit admission following an anesthesia-related intraoperative complication was performed. Secondary outcomes included anesthesia factors associated with an anesthesia-related intraoperative event, and postoperative complications and surgical outcomes between patients admitted to the intensive care unit and those who were not.Two hundred ninety-six patients with Chiari I Malformation were identified. Clinical characteristics associated with an unanticipated admission to the intensive care unit were younger age, American Society of Anesthesiologist (ASA) physical status2 and an anesthesia-related intraoperative complication. 29 anesthesia-related intraoperative complications were observed in 25 patients (8.4%). Two of 25 patients (8%) with an anesthesia-related intraoperative complication compared with 3 of 271 (1%) patients without anesthesia-related intraoperative complication had an unanticipated admission to the intensive care unit, odds ratio 7.8 (95% CI 1.2-48.8, p = .010). When adjusted for age, sex, ASA physical status, presenting symptoms, concomitant syringomyelia, previous decompression surgery and year of surgery, the odds ratio for an unanticipated admission to the intensive care unit following an anesthesia-related intraoperative complication was 5.9 (95% CI 0.51-59.6, p = .149). There were no differences in surgical outcomes between patients with or without an unanticipated admission to the intensive care unit.Our study demonstrates that although anesthesia-related intraoperative complications during posterior fossa decompression are infrequent, they are associated with an increased risk of an unanticipated admission to the intensive care unit.
- Published
- 2022
9. HGG-02. Epigenetic transcription regulation and 3D genome structure in pediatric high-grade glioma
- Author
-
Tina Huang, Juan Wang, Ye Hu, Andrea Piunti, Elizabeth Bartom, Ali Shilatifard, Feng Yue, and Amanda Saratsis
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
INTRODUCTION: Pediatric high-grade gliomas (pHGGs), including glioblastoma multiforme (GBM) and diffuse intrinsic pontine glioma (DIPG), are highly morbid brain tumors. Up to 80% of DIPGs harbor a somatic missense mutation in genes encoding Histone H3. To investigate whether the H3K27M mutant protein is associated with distinct chromatin structure affecting transcription regulation, we generated the first high-resolution Hi-C and ATAC-Seq maps of pHGG cell lines, and integrated these with tissue and cell genomic data. METHODS: We generated sequencing data from patient-derived cell lines (DIPG n=6, GBM n=3, normal n=2) and frozen tissue specimens (DIPG n=1, normal brainstem n=1). Analyses included cell line RNA-Seq, ChIP-Seq (H3K27ac, H3K27me3, H3K27M) and genome-wide chromatin conformation capture (Hi-C), as well as tissue ATAC-Seq. Publicly available pediatric glioma tissue ChIP-Seq data was integrated with cell data. CRISPR knock-down of target enhancer regions was performed. RESULTS: We identified tumor-specific enhancers and regulatory networks for known oncogenes in DIPG and GBM. In DIPG, FOX, SOX, STAT and SMAD families were among top H3K27Ac enriched motifs. Significant differences in Topologically Associating Domains (TADs) and DNA looping were observed at OLIG2 and MYCN in H3K27M mutant DIPG, relative to wild-type GBM and normal cells. Pharmacologic treatment targeting H3K27Ac (BET and Bromodomain inhibition) altered these 3D structures. Functional analysis of differentially enriched enhancers in DIPG implicated SOX2, SUZ12, and TRIM24 as top activated upstream regulators. Distinct genomic structural variations leading to enhancer hijacking and gene co-amplification were identified at A2M, JAG2, and FLRT1. CONCLUSION: We show genome structural variations enhancer-promoter interactions that impact gene expression in pHGG in the presence and absence of the H3K27M mutation. Our results imply that tridimensional genome alterations may play a critical role in the pHGG epigenetic landscape and thereby contribute to pediatric gliomagenesis. Further studies examining the impact of the alterations are therefore underway.
- Published
- 2022
10. DIPG-49. International preclinical drug discovery and biomarker program informing an adoptive combinatorial trial for DMG
- Author
-
Javad Nazarian, Matthew Dun, Lindsay Kilburn, Sebastian Waszak, Nicholas Vitanza, Andrea Franson, Mike Prados, Eric Raabe, Ron Firestein, Alexander Beck, Amanda Saratsis, Barak Rotblat, Dannis van Vuurder, Jessica Foster, Esther Hulleman, Cassie Kline, Nalin Gupta, Jason Cain, Carl Koschmann, and Sabine Muller
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
INTRODUCTION: DMG-ACT (DMG- multi-arm Adaptive and Combinatorial Trial) will implement an innovative clinical trial design of combinatorial arms for patients with DMG at all disease stages, that is adaptive to pre-clinical and correlate data generated in eight collaborating institutions. The goal of the team is to rapidly identify and validate i) promising drugs and drug combinations for clinical use, and ii) predictive biomarkers of promising drugs. METHODS: In vitro (n=30) and in vivo (n=8) models of DMG across fourteen institutions were used to assess single and combination treatment of over 80 drugs and drug combinations. Predictive biomarkers of response for top candidate drugs were identified using extensive molecular assays including proteomics, CRISPR, RNAseq, ELISA, FACS, and IHC. RESULTS: Inhibitory concentration (IC50) of all drugs were established and validated across all participating sites. In vivo validation of single and combination drug assays confirmed drug efficacy as increased survival for: ONC201 (p=0.01), ONC206 (p=0.01), ONC201+ONC206 (p=0.02), ONC201+panobinostat (p=0.01). Marizomib was highly toxic in murine PDX and zebrafish larvae assays. Murine pharmacokinetic analysis showed peak brain levels of ONC201, and ONC206 above pre-clinical IC50 concentrations. Molecular testing and analyses of existing drug screen across 578 cancer cells validated mitochondrial stress and additional proteins, as the main targets induces by ONC201/6. CONCLUSION: Thorough preclinical testing in a multi-site laboratory setting identified promising therapeutics for DMGs, resulting in launch of two clinical trials (PNOC022, ONOC023). Validation of identified biomarkers are ongoing using clinical specimen as well as in vivo PDX models.
- Published
- 2022
11. Cerebrospinal Fluid Access Devices in Pediatric Diffuse Midline Glioma Patients: Literature Review and Evaluation of Institutional Practices
- Author
-
Daphne Li, Wendy Stellpflug, and Amanda Saratsis
- Subjects
medicine.medical_specialty ,Cerebrospinal fluid ,business.industry ,Glioma ,medicine ,Surgery ,Neurology (clinical) ,Radiology ,business ,medicine.disease - Published
- 2019
12. Choroidal artery embolization in the management of cerebrospinal fluid overproduction: case report and review of the literature
- Author
-
Brian D. Rothstein, Tahaamin Shokuhfar, Ali Shaibani, Amanda Saratsis, Tord D. Alden, Daphne Li, and Julia Pantalone
- Subjects
medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Endoscopic third ventriculostomy ,Macrocephaly ,General Medicine ,medicine.disease ,Hydrocephalus ,Shunting ,Anterior choroidal artery ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Medicine ,Choroid plexus ,030212 general & internal medicine ,Radiology ,Embolization ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Diffuse villous hyperplasia of the choroid plexus (DVHCP) is a rare cause of communicating hydrocephalus. DVHCP may be diagnosed radiographically and through histological evaluation. It may be associated with genetic abnormalities, particularly involving chromosome 9. Due to CSF overproduction, patients with DVHCP often fail management with shunting alone and may require adjuvant interventions. The authors present the case of a child with partial trisomy 9p and delayed diagnosis of hydrocephalus with radiographic evidence of DVHCP who was successfully managed with ventriculoperitoneal shunt (VPS) placement, adjuvant bilateral endoscopic choroid plexus coagulation (CPC), and the novel application of anterior choroidal artery embolization. In addition, a systematic MEDLINE search was conducted using the keywords “diffuse villous hyperplasia,” “choroid plexus hypertrophy,” and “idiopathic cerebrospinal fluid overproduction.” Clinicopathological characteristics and outcomes of the present case were reviewed and compared to those in the literature.A 14-month-old girl with partial trisomy 9p presented with macrocephaly and radiographic evidence of communicating hydrocephalus and DVHCP. Ventriculoperitoneal shunting resulted in distal failure due to inadequate CSF absorption, and ventriculoatrial shunt (VAS) placement was not possible due to multiple cardiac anomalies. Daily CSF production was reduced via endoscopic third ventriculostomy and bilateral CPC, followed by distal choroidal artery embolization, enabling VPS re-internalization. The embolization was complicated by radiographic evidence of an iatrogenic cerebral infarct, but this was clinically occult. Thirty-two additional cases of communicating hydrocephalus due to DVHCP are reported in the literature: 27 pediatric, 3 adult, and 2 postmortem. Genetic abnormalities were noted in 14, with 7 (50%) involving chromosome 9. Twelve patients underwent plexectomy (9 bilateral, 2 unilateral, 1 partial), and 10 underwent CPC (4 bilateral, 3 unilateral, and 3 unspecified), with or without shunting. Eight patients were successfully managed with shunting alone (6 VASs, 2 VPSs), and none underwent arterial embolization.DVHCP is a rare cause of communicating hydrocephalus that may be associated with genetic abnormalities. A thorough review of the literature highlights diagnostic criteria and interventional options involved in managing this cause of CSF overproduction. The present case demonstrates that angiographic confirmation of prominent choroidal arteries may contribute to the diagnosis DVHCP. In addition, embolization of the distal choroidal arteries may be considered as a potential adjuvant treatment in patients for whom conventional treatments have failed or are not feasible.
- Published
- 2019
13. Standardization of the liquid biopsy for pediatric diffuse midline glioma using ddPCR
- Author
-
Carl Koschmann, Sabine Mueller, Eshini Panditharatna, Amanda Saratsis, Erin R Bonner, Daphne Li, Tina Huang, Javad Nazarian, Rishi Lulla, Kyle Wierzbicki, University of Zurich, Nazarian, Javad, and Saratsis, Amanda M
- Subjects
Poor prognosis ,Science ,610 Medicine & health ,medicine.disease_cause ,Polymerase Chain Reaction ,Sensitivity and Specificity ,Article ,Circulating Tumor DNA ,Histones ,Tumour biomarkers ,Mutation Rate ,Glioma ,Biomarkers, Tumor ,medicine ,Humans ,Liquid biopsy ,Child ,Allele frequency ,Cancer genetics ,Mutation ,1000 Multidisciplinary ,Multidisciplinary ,Brain Neoplasms ,business.industry ,Liquid Biopsy ,Reproducibility of Results ,Reference Standards ,Prognosis ,medicine.disease ,Molecular biology ,Primary tumor ,Subtyping ,Therapeutic monitoring ,CNS cancer ,10036 Medical Clinic ,Feasibility Studies ,Medicine ,business - Abstract
Diffuse midline glioma (DMG) is a highly morbid pediatric brain tumor. Up to 80% of DMGs harbor mutations in histone H3-encoding genes, associated with poor prognosis. We previously showed the feasibility of detecting H3 mutations in circulating tumor DNA (ctDNA) in the liquid biome of children diagnosed with DMG. However, detection of low levels of ctDNA is highly dependent on platform sensitivity and sample type. To address this, we optimized ctDNA detection sensitivity and specificity across two commonly used digital droplet PCR (ddPCR) platforms (RainDance and BioRad), and validated methods for detecting H3F3A c.83A > T (H3.3K27M) mutations in DMG CSF, plasma, and primary tumor specimens across three different institutions. DNA was extracted from H3.3K27M mutant and H3 wildtype (H3WT) specimens, including H3.3K27M tumor tissue (n = 4), CSF (n = 6), plasma (n = 4), and human primary pediatric glioma cells (H3.3K27M, n = 2; H3WT, n = 1). ctDNA detection was enhanced via PCR pre-amplification and use of distinct custom primers and fluorescent LNA probes for c.83 A > T H3F3A mutation detection. Mutation allelic frequency (MAF) was determined and validated through parallel analysis of matched H3.3K27M tissue specimens (n = 3). We determined technical nuances between ddPCR instruments, and optimized sample preparation and sequencing protocols for H3.3K27M mutation detection and quantification. We observed 100% sensitivity and specificity for mutation detection in matched DMG tissue and CSF across assays, platforms and institutions. ctDNA is reliably and reproducibly detected in the liquid biome using ddPCR, representing a clinically feasible, reproducible, and minimally invasive approach for DMG diagnosis, molecular subtyping and therapeutic monitoring.
- Published
- 2021
14. Epigenomic landscape and 3D genome structure in pediatric high-grade glioma
- Author
-
Juan Wang, Ye Hou, Ali Shilatifard, Xinyan Lu, Tina Yi-Ting Huang, Feng Yue, Elizabeth T. Bartom, and Amanda Saratsis
- Subjects
Epigenomics ,0303 health sciences ,Multidisciplinary ,Glioma ,Biology ,medicine.disease_cause ,Genome ,Chromatin ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,medicine ,Brain Stem Neoplasms ,Humans ,Epigenetics ,Carcinogenesis ,Enhancer ,Child ,Gene ,030304 developmental biology - Abstract
Pediatric high-grade gliomas (pHGGs), including glioblastoma multiforme (GBM) and diffuse intrinsic pontine glioma (DIPG), are morbid brain tumors. Even with treatment survival is poor, making pHGG the number one cause of cancer death in children. Up to 80% of DIPGs harbor a somatic missense mutation in genes encoding histone H3. To investigate whether H3K27M is associated with distinct chromatin structure that alters transcription regulation, we generated the first high-resolution Hi-C maps of pHGG cell lines and tumor tissue. By integrating transcriptome (RNA-seq), enhancer landscape (ChIP-seq), genome structure (Hi-C), and chromatin accessibility (ATAC-seq) datasets from H3K27M and wild-type specimens, we identified tumor-specific enhancers and regulatory networks for known oncogenes. We identified genomic structural variations that lead to potential enhancer hijacking and gene coamplification, including A2M, JAG2, and FLRT1 Together, our results imply three-dimensional genome alterations may play a critical role in the pHGG epigenetic landscape and contribute to tumorigenesis.
- Published
- 2021
15. Mesenchymal Stem Cells Successfully Deliver Oncolytic Virotherapy to Diffuse Intrinsic Pontine Glioma
- Author
-
Ting Xiao, Markella Zannikou, Amanda Saratsis, Yu Han, Irina V. Balyasnikova, Erin R. Bonner, Adam M. Sonabend, Michael Chastkofsky, Javad Nazarian, C. David James, Rintaro Hashizume, David T. Curiel, Maciej S. Lesniak, Katarzyna C. Pituch, Hiroaki Katagi, Liliana Ilut, Craig Horbinski, and University of Zurich
- Subjects
0301 basic medicine ,Cancer Research ,Adolescent ,medicine.medical_treatment ,Mice, Nude ,Context (language use) ,610 Medicine & health ,Apoptosis ,Cell Surface Proteins ,Mesenchymal Stem Cell Transplantation ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Viral entry ,Survivin ,medicine ,Tumor Cells, Cultured ,Animals ,Brain Stem Neoplasms ,Humans ,Promoter Regions, Genetic ,Cell Proliferation ,Oncolytic Virotherapy ,Mice, Inbred BALB C ,business.industry ,Mesenchymal stem cell ,Diffuse Intrinsic Pontine Glioma ,Mesenchymal Stem Cells ,Prognosis ,Xenograft Model Antitumor Assays ,Oncolytic virus ,Radiation therapy ,Oncolytic Viruses ,030104 developmental biology ,Oncology ,Cell culture ,10036 Medical Clinic ,030220 oncology & carcinogenesis ,Cancer research ,Female ,business - Abstract
Purpose: Diffuse intrinsic pontine glioma (DIPG) is among the deadliest of pediatric brain tumors. Radiotherapy is the standard-of-care treatment for DIPG, but offers only transient relief of symptoms for patients with DIPG without providing significant survival benefit. Oncolytic virotherapy is an anticancer treatment that has been investigated for treating various types of brain tumors. Experimental Design: Here, we have explored the use of mesenchymal stem cells (MSC) for oncolytic virus (OV) delivery and evaluated treatment efficacy using preclinical models of DIPG. The survivin promoter drives the conditional replication of OV used in our studies. The efficiency of OV entry into the cells is mediated by fiber modification with seven lysine residues (CRAd.S.pK7). Patients' samples and cell lines were analyzed for the expression of viral entry proteins and survivin. The ability of MSCs to deliver OV to DIPG was studied in the context of a low dose of irradiation. Results: Our results show that DIPG cells and tumors exhibit robust expression of cell surface proteins and survivin that enable efficient OV entry and replication in DIPG cells. MSCs loaded with OV disseminate within a tumor and release OV throughout the DIPG brainstem xenografts in mice. Administration of OV-loaded MSCs with radiotherapy to mice bearing brainstem DIPG xenografts results in more prolonged survival relative to that conferred by either therapy alone (P < 0.01). Conclusions: Our study supports OV, CRAd.S.pK7, encapsulated within MSCs as a therapeutic strategy that merits further investigation and potential translation for DIPG treatment.
- Published
- 2020
16. Effects of H3.3G34V mutation on genomic H3K36 and H3K27 methylation patterns in isogenic pediatric glioma cells
- Author
-
Ali Shilatifard, Jin Qi, Elizabeth T. Bartom, Marc A. Morgan, Tina Yi-Ting Huang, Amanda Saratsis, and Andrea Piunti
- Subjects
Chromatin Immunoprecipitation ,Cell Survival ,Mutant ,Mutation, Missense ,medicine.disease_cause ,Methylation ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,Histones ,Cellular and Molecular Neuroscience ,Mutant protein ,Cell Line, Tumor ,Gene expression ,medicine ,Humans ,Child ,lcsh:Neurology. Diseases of the nervous system ,Cell Proliferation ,Mutation ,Gene knockdown ,Histone H3 mutations ,biology ,Brain Neoplasms ,Research ,H3K36me3 ,High-Throughput Nucleotide Sequencing ,Glioma ,Molecular biology ,Isogenic human disease models ,Gene Expression Regulation, Neoplastic ,Histone Code ,Histone ,Astrocytes ,Gene Knockdown Techniques ,Pediatric high-grade glioma ,biology.protein ,Chromatin Immunoprecipitation Sequencing ,Neurology (clinical) ,Carcinogenesis ,Post-translational modifications - Abstract
Histone H3.3 mutation (H3F3A) occurs in 50% of cortical pediatric high-grade gliomas. This mutation replaces glycine 34 with arginine or valine (G34R/V), impairing SETD2 activity (H3K36-specific trimethyltransferase). Consequently, reduced H3K36me3 is observed on H3.3G34V nucleosomes relative to wild-type, contributing to genomic instability and driving a distinct gene expression signature associated with tumorigenesis. However, it is not known if this differential H3K36me3 enrichment is due to H3.3G34V mutant protein alone. Therefore, we set to elucidate the effect of H3.3G34V mutant protein in pediatric glioma on H3K36me3, H3K27me3 and H3.3 enrichment in vitro. We found that the doxycycline-inducible shRNA knockdown of mutant H3F3A encoding the H3.3G34V protein resulted in loss of H3.3G34V enrichment and increased H3K36me3 enrichment throughout the genome. After knockdown, H3.3G34V enrichment was preserved at loci observed to have the greatest H3.3G34V and H3K36me3 enrichment prior to knockdown. Induced expression of mutant H3.3G34V protein in vitro was insufficient to induce genomic H3K36me3 enrichment patterns observed in H3.3G34V mutant glioma cells. We also observed strong co-enrichment of H3.3G34V and wild-type H3.3 protein, as well as greater H3K27me3 enrichment, in cells expressing H3.3G34V. Taken together, our study demonstrates the effects of H3.3G34V mutant protein on genomic H3K36me3, H3K27me3 and H3.3 enrichment patterns in isogenic cell lines.
- Published
- 2020
17. Histone tail analysis reveals H3K36me2 and H4K16ac as epigenetic signatures of diffuse intrinsic pontine glioma
- Author
-
Juliette A. Morris, Javad Nazarian, Paul M. Thomas, Daphne Li, Amanda Saratsis, Madeline A. Zoltek, Jeannie M. Camarillo, Jin Qi, Shejuan An, Mandana Behbahani, Neil L. Kelleher, Tina Yi Ting Huang, Madhuri Kambhampati, University of Zurich, and Saratsis, Amanda M
- Subjects
0301 basic medicine ,Epigenomics ,Male ,Cancer Research ,Mutant ,610 Medicine & health ,Histones ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,H3K27M ,Brain Stem Neoplasms ,Humans ,1306 Cancer Research ,Epigenetics ,biology ,Chemistry ,Research ,Gene Expression Profiling ,Diffuse Intrinsic Pontine Glioma ,H3K36me2 ,Bromodomain ,Histone ,030104 developmental biology ,Targeted mass spectrometry ,Oncology ,Acetylation ,Cell culture ,10036 Medical Clinic ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,DIPG ,Female ,2730 Oncology ,H4K16ac - Abstract
Background Diffuse intrinsic pontine glioma (DIPG) is an aggressive pediatric brainstem tumor. Most DIPGs harbor a histone H3 mutation, which alters histone post-translational modification (PTM) states and transcription. Here, we employed quantitative proteomic analysis to elucidate the impact of the H3.3K27M mutation, as well as radiation and bromodomain inhibition (BRDi) with JQ1, on DIPG PTM profiles. Methods We performed targeted mass spectrometry on H3.3K27M mutant and wild-type tissues (n = 12) and cell lines (n = 7). Results We found 29.2 and 26.4% of total H3.3K27 peptides were H3.3K27M in mutant DIPG tumor cell lines and tissue specimens, respectively. Significant differences in modification states were observed in H3.3K27M specimens, including at H3K27, H3K36, and H4K16. In addition, H3.3K27me1 and H4K16ac were the most significantly distinct modifications in H3.3K27M mutant tumors, relative to wild-type. Further, H3.3K36me2 was the most abundant co-occurring modification on the H3.3K27M mutant peptide in DIPG tissue, while H4K16ac was the most acetylated residue. Radiation treatment caused changes in PTM abundance in vitro, including increased H3K9me3. JQ1 treatment resulted in increased mono- and di-methylation of H3.1K27, H3.3K27, H3.3K36 and H4K20 in vitro. Conclusion Taken together, our findings provide insight into the effects of the H3K27M mutation on histone modification states and response to treatment, and suggest that H3K36me2 and H4K16ac may represent unique tumor epigenetic signatures for targeted DIPG therapy.
- Published
- 2020
18. MSCs Successfully Deliver Oncolytic Virotherapy to Diffuse Intrinsic Pontine Glioma
- Author
-
Ting Xiao, Liliana Ilut, Michael Chastkofsky, Javad Nazarian, Craig Horbinski, Erin R. Bonner, Amanda Saratsis, Maciej S. Lesniak, Yu Han, David T. Curiel, C. D. James, Rintaro Hashizume, Katarzyna C. Pituch, Balyasnikova, Hiroaki Katagi, and Adam M. Sonabend
- Subjects
0303 health sciences ,Standard of care ,business.industry ,medicine.medical_treatment ,Mesenchymal stem cell ,Cell Surface Proteins ,Treatment efficacy ,3. Good health ,Oncolytic virus ,Radiation therapy ,03 medical and health sciences ,0302 clinical medicine ,Pediatric brain ,Anticancer treatment ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,business ,030304 developmental biology - Abstract
Diffuse intrinsic pontine glioma (DIPG) is among the deadliest of pediatric brain tumors. Radiation therapy is the standard of care treatment for DIPG, but offers only transient relief of symptoms for DIPG patients without providing significant survival benefit. Oncolytic virotherapy (OV) is an anticancer treatment that has been investigated for treating various types of brain tumors. Here, we have explored the use of mesenchymal stem cells (MSC) for OV delivery and evaluated treatment efficacy using preclinical models of DIPG. Our results show that DIPG cells and tumors exhibit robust expression of cell surface proteins that are important for OV entry, and that MSCs loaded with OV disseminate within and release OV throughout the tumor in mice bearing DIPG brainstem xenografts. When combining administration of OV-loaded MSCs with radiotherapy, mice bearing brainstem DIPG xenografts experience a significant survival benefit, relative to that conferred by either therapy alone (p
- Published
- 2020
- Full Text
- View/download PDF
19. EPCO-20. PEDIATRIC HIGH-GRADE GLIOMA EXHIBITS DISTINCT 3D GENOME STRUCTURE THAT IMPACTS TRANSCRIPTION REGULATION
- Author
-
Juan Wang, Elizabeth T. Bartom, Tina Huang, Amanda Saratsis, Ali Shilatifard, Feng Yue, Ye Hu, and Andrea Piunti
- Subjects
Genetics ,Cancer Research ,Mutation ,RNA-Seq ,Biology ,medicine.disease ,medicine.disease_cause ,Genome ,Oncology ,Glioma ,Gene expression ,medicine ,Transcriptional regulation ,Neurology (clinical) ,Epigenetics ,Gene - Abstract
INTRODUCTION Pediatric high-grade gliomas (pHGGs), including glioblastoma multiforme (GBM) and diffuse intrinsic pontine glioma (DIPG), are highly morbid brain tumors. Up to 80% of DIPGs harbor a somatic missense mutation in genes encoding Histone H3. To investigate whether the H3K27M mutant protein is associated with distinct chromatin structure affecting transcription regulation, we generated the first high-resolution Hi-C and ATAC-Seq maps of pHGG cell lines, and integrated these with tissue and cell genomic data. METHODS We generated sequencing data from patient-derived cell lines (DIPG n=6, GBM n=3, normal n=2) and frozen tissue specimens (DIPG n=1, normal brainstem n=1). Analyses included cell line RNA-Seq, ChIP-Seq (H3K27ac, H3K27me3, H3K27M) and genome-wide chromatin conformation capture (Hi-C), as well as tissue ATAC-Seq. Publicly available pediatric glioma tissue ChIP-Seq data was integrated with cell data. CRISPR knock-down of target enhancer regions was performed. RESULTS We identified tumor-specific enhancers and regulatory networks for known oncogenes in DIPG and GBM. In DIPG, FOX, SOX, STAT and SMAD families were among top H3K27Ac enriched motifs. Significant differences in Topologically Associating Domains (TADs) and DNA looping were observed at OLIG2 and MYCN in H3K27M mutant DIPG, relative to wild-type GBM and normal cells. Pharmacologic treatment targeting H3K27Ac (BET and Bromodomain inhibition) altered these 3D structures. Functional analysis of differentially enriched enhancers in DIPG implicated SOX2, SUZ12, and TRIM24 as top activated upstream regulators. Distinct genomic structural variations leading to enhancer hijacking and gene co-amplification were identified at A2M, JAG2, and FLRT1. CONCLUSION We show genome structural variations enhancer-promoter interactions that impact gene expression in pHGG in the presence and absence of the H3K27M mutation. Our results imply that tridimensional genome alterations may play a critical role in the pHGG epigenetic landscape and thereby contribute to pediatric gliomagenesis. Further studies examining the impact of the alterations is therefore underway.
- Published
- 2021
20. HGG-01. 3D GENOME STRUCTURE IMPACTS GENE EXPRESSION IN PEDIATRIC HIGH-GRADE GLIOMA
- Author
-
Juan Wang, Elizabeth T. Bartom, Tina Huang, Amanda Saratsis, Ye Hou, Andrea Piunti, Ali Shilatifard, and Feng Yue
- Subjects
Cancer Research ,RNA-Seq ,Biology ,Genome structure ,Genome ,Oncology ,Gene expression ,Cancer research ,Transcriptional regulation ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,High Grade Gliomas ,Neurology (clinical) ,Epigenetics ,Gene ,High-Grade Glioma - Abstract
Introduction Pediatric high-grade gliomas (pHGGs), including glioblastoma multiforme (GBM) and diffuse intrinsic pontine glioma (DIPG), are highly morbid brain tumors. Up to 80% of DIPGs harbor a somatic missense mutation in genes encoding Histone H3. To investigate whether the H3K27M mutant protein is associated with distinct chromatin structure affecting transcription regulation, we generated the first high-resolution Hi-C and ATAC-Seq maps of pHGG cell lines, and integrated these with tissue and cell genomic data. Methods We generated sequencing data from patient-derived cell lines (DIPG n=6, GBM n=3, normal n=2) and frozen tissue specimens (DIPG n=1, normal brainstem n=1). Analyses included cell line RNA-Seq, ChIP-Seq (H3K27ac, H3K27me3, H3K27M) and genome-wide chromatin conformation capture (Hi-C), as well as tissue ATAC-Seq. Publicly available pediatric glioma tissue ChIP-Seq data was integrated with cell data. Results We identified tumor-specific enhancers and regulatory networks for known oncogenes in DIPG and GBM. In DIPG, FOX, SOX, STAT and SMAD families were among top H3K27Ac enriched motifs. Significant differences in Topologically Associating Domains (TADs) and DNA looping were observed at OLIG2 and MYCN in H3K27M mutant DIPG, relative to wild-type GBM and normal cells. Pharmacologic treatment targeting H3K27Ac (BET and Bromodomain inhibition) altered these 3D structures. Functional analysis of differentially enriched enhancers in DIPG implicated SOX2, SUZ12, and TRIM24 as top activated upstream regulators. Distinct genomic structural variations leading to enhancer hijacking and gene co-amplification were identified at A2M, JAG2, and FLRT1. Conclusion We show genome structural variations enhancer-promoter interactions that impact gene expression in pHGG in the presence and absence of the H3K27M mutation. Our results imply that tridimensional genome alterations may play a critical role in the pHGG epigenetic landscape and thereby contribute to pediatric gliomagenesis. Further studies examining the impact of the alterations, including CRISPR knock-down of target enhancer regions, is therefore underway.
- Published
- 2021
21. PDTM-31. INVESTIGATING THE EFFECT OF H3G34V MUTATION ON DISTINCT GENOMIC H3K36 METHYLATION PATTERNS IN PEDIATRIC GLIOMA
- Author
-
Elizabeth Barton, Rintaro Hashizume, Jin Qi, Andrea Piunti, Ali Shilatifard, Tina Huang, Amanda Saratsis, and Patrick A. Ozark
- Subjects
Genome instability ,Genetics ,Cancer Research ,Mutation ,Pediatric Tumors ,Methylation ,Biology ,medicine.disease_cause ,medicine.disease ,Genome ,Oncology ,Glioma ,medicine ,Neurology (clinical) ,Carcinogenesis ,Allele frequency ,Gene - Abstract
BACKGROUND Histone H3.3 mutation (H3F3A) occurs in 50% of cortical pediatric high-grade gliomas (pHGGs). This mutation replaces glycine 34 with arginine or valine (G34R/V), impairing SETD2 activity (H3K36-specific trimethyltransferase). Consequently, reduced H3K36me is observed on H3G34V nucleosomes relative to wild-type, contributing to genomic instability and driving a distinct gene expression signature associated with tumorigenesis. However, it is not known if this differential H3K36me3 enrichment is due to H3G34V mutant protein alone. Therefore, we set to elucidate the effect of H3G34V mutation in pediatric glioma on genomic H3K36me3 enrichment in vitro. METHODS Doxycycline-inducible short hairpin RNA (shRNA) against H3F3A was delivered via lentivirus to established H3G34V mutant pediatric glioma cell line KNS42, and H3G34V introduced into H3.3 wild type normal human astrocytes (NHA). Transfections were confirmed by western blot, fluorescent imaging, and flow cytometry, with resulting H3.3WT and H3K36me3 expression determined by western blot. H3.3WT, H3K36me3, and H3G34V ChIP-Seq was performed to evaluate effects of H3G34V mutation on genomic enrichment patterns. RESULTS Complete knockdown of H3G34V was achieved with DOX-induced shRNA, with no change in total H3.3, suggesting disproportionate allelic frequency of genes encoding H3.3 (H3F3A and H3F3B). Modest increase in H3K36me3 expression occurred after H3F3A-knockdown from KNS42, suggesting H3G34V alone impacts observed H3K36me3 levels. H3G34V knock-in to NHAs was verified via western blot. Distinct H3K36me3 genomic enrichment was observed with H3G34V on ChIP-Seq. CONCLUSIONS We demonstrate that DOX-inducible knockdown of H3F3A in an H3G34V mutant pediatric glioma cell line, and H3G34V mutation transduction in wild-type NHAs, directly impacts H3K36me3 expression level. Further evaluation by ChIP-Seq analysis for restoration of wild-type genomic H3K36me3 enrichment patterns with H3G34V knockdown, and mutant H3K36me3 patterns with H3G34V transduction, is currently underway.
- Published
- 2019
22. Tenascin-C expression contributes to pediatric brainstem glioma tumor phenotype and represents a novel biomarker of disease
- Author
-
Amanda Saratsis, Patrick A. Ozark, S. An, Tina Huang, Jin Qi, Erin R. Bonner, Elizabeth T. Bartom, Craig Horbinski, Rintaro Hashizume, Charles David James, and Darian R. Esfahani
- Subjects
Male ,0301 basic medicine ,Tenascin-C ,Cell morphology ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Diffuse intrinsic pontine glioma (DIPG) ,Histone H3 mutation (H3K27 M) ,Cell Line, Tumor ,Glioma ,Biomarkers, Tumor ,medicine ,Brain Stem Neoplasms ,Humans ,Progression-free survival ,Child ,lcsh:Neurology. Diseases of the nervous system ,Gene knockdown ,biology ,Cell growth ,Research ,Tenascin C ,Diffuse midline glioma ,Tenascin ,Cell migration ,musculoskeletal system ,medicine.disease ,3. Good health ,030104 developmental biology ,Child, Preschool ,biology.protein ,Cancer research ,Biomarker (medicine) ,Female ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
Diffuse intrinsic pontine glioma (DIPG), an infiltrative, high grade glioma (HGG) affecting young children, has the highest mortality rate of all pediatric cancers. Despite treatment, average survival is less than twelve months, and five-year survival under 5%. We previously detected increased expression of Tenascin-C (TNC) protein in DIPG cerebrospinal fluid and tumor tissue relative to normal specimens. TNC is an extracellular matrix (ECM) glycoprotein that mediates cell-matrix interactions, guides migrating neurons during normal brain development and is thought to maintain the periventricular stem cell niche in the developing brain. Tumor TNC expression is reported in adult glioma and other cancers. However, the pattern and effects of TNC expression in DIPG has not been previously explored. Here, we characterize TNC expression in patient derived pediatric supratentorial HGG (n = 3) and DIPG (n = 6) cell lines, as well as pediatric glioma tumor (n = 50) and normal brain tissue specimens (n = 3). We found tumor specific TNC gene and protein overexpression that directly correlated with higher tumor grade (WHO III and IV, p = 0.05), H3K27 M mutation (p = 0.012), shorter progression free survival (p = 0.034), and poorer overall survival (0.041) in association with these factors. TNC knockdown via lentiviral shRNA transfection of HGG (n = 1) and DIPG (n = 3) cell lines resulted in decreased cell proliferation, migration, and invasion in vitro (p
- Published
- 2019
23. HGG-10. HISTONE H3G34V MUTATION IS SUFFICIENT TO DRIVE DISTINCT GENOMIC H3K36 METHYLATION PATTERNS IN PEDIATRIC GLIOMA
- Author
-
Jin Qi, Elizabeth T. Bartom, Ali Shilatifard, Patrick A. Ozark, Tina Huang, Amanda Saratsis, Rintaro Hashizume, and Andrea Piunti
- Subjects
Cancer Research ,Mutation ,biology ,Point mutation ,Methylation ,medicine.disease_cause ,medicine.disease ,Histone ,Oncology ,Glioma ,biology.protein ,Cancer research ,Transcriptional regulation ,medicine ,Nucleosome ,Neurology (clinical) ,Carcinogenesis ,High Grade Glioma - Abstract
BACKGROUND: Mutations in genes encoding Histone protein isoforms H3.3 (H3F3A and H3F3A) and H3.1 (HIST1H3B) are detected at high frequency in pediatric high-grade glioma (pHGG). Up to 50% of cortical pHGGs bear a point mutation in H3F3A, resulting in replacement of glycine 34 with arginine or valine (G34R/V). H3G34V mutation is known to impair SETD2 activity (H3K36-specific trimethyltransferase). Reduced K36me3 is observed on nucleosomes bearing H3G34V protein relative to wild-type, contributing to genomic instability and driving a distinct gene expression signature associated with tumorigenesis. However, it is not known if this differential H3K36me3 enrichment is due to H3G34V mutant protein alone. We therefore set to elucidate the effect of H3G34V mutant protein on genomic H3K36me3 enrichment in vitro. METHODS: A doxycycline-inducible short hairpin RNA (shRNA) against G34V mutant H3F3A was delivered via lentivirus to an established G34V mutant pediatric glioma cell line (KNS42). H3G34V mutation was introduced into H3.3 wild type normal human astrocytes (NHA). Mutation knockdown / knockin was confirmed by sequencing and western blot. H3.3WT, H3K36me3, and H3G34V ChIP-Seq was performed to determine relative genomic distribution in transfected and unperturbed KNS42 cells and NHAs, and also compared to a H3WT glioma cell line. RESULTS: shRNA-induced knockdown of H3G34V restored the pattern of genomic H3K36me3 enrichment to that observed in H3.3 wild-type glioma cells and NHAs. In turn, introducing H3G34V to NHAs was sufficient to drive the distinct K36 methylation profile seen in H3G34V glioma cells. H3G34V mutation was also associated with a global increase in overall H3.3WT genomic enrichment, representing a heightened state of active transcription relative to wild-types. CONCLUSIONS: Our study is the first to demonstrate H3G34V mutation is sufficient to induce differential H3K36me3 enrichment patterns associated with genomic instability and increased transcription. Gene expression analysis is currently underway to further evaluate these effects on transcription regulation.
- Published
- 2019
24. DIPG-39. NOVEL PROTEOMIC ANALYSIS REVEALS EPIGENETIC THERAPEUTIC TARGETS IN PEDIATRIC GLIOMA
- Author
-
Tina Huang, Jeannie M. Camarillo, She-Juan An, Daphne Li, Javad Nazarian, Andrea Piunti, Paul M. Thomas, Jin Qi, and Amanda Saratsis
- Subjects
Cancer Research ,business.industry ,Diffuse Midline Glioma/DIPG ,Childhood cancer ,medicine.disease_cause ,Proteomics ,medicine.disease ,Oncology ,Glioma ,Pediatric glioma ,medicine ,Cancer research ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Epigenetics ,Carcinogenesis ,business - Abstract
INTRODUCTION Diffuse midline glioma is a highly morbid pediatric cancer. Up to 80% harbor Histone H3K27M mutation, which alters Histone H3 post-translational modifications (PTMs) and genomic enrichment patterns, affecting chromatin structure and transcription. We previously identified tumorigenic patterns of H3K27Ac/bromodomain co-enrichment and pre-clinical efficacy of bromodomain inhibition (JQ1) in DMG. Here, we employ a novel proteomics approach developed at our institution to further elucidate the impact of H3K27M mutation on glioma epigenetic signatures and treatment response. METHODS Epiproteomic analysis was performed on pediatric glioma cells (H3K27 WT n=2, H3K27M n=2) to characterize 95 distinct Histone H3 N-terminal tail modification states. Cells were treated with JQ1 or DMSO, and collected at 0h, 24h, 48h, Histones extracted from isolated nuclei and immunopurified, then analyzed by LC-MS/MS. Results were integrated with RNA-Seq and ChIP Seq (H3.3K27M, H3.3, H3K27Ac, H3K27me3, H3K4me1, H3K4me3) from the same cell lines. Pediatric glioma tissues (H3K27M WT n=3, H3K27M n= 9) were similarly analyzed to validate cell line results. RESULTS Cell PTM profiles cluster by H3 mutation status on unsupervised analysis. Significant differential PTM abundance and genomic enrichment H3K27M, H3.3 WT, H3K27Me3 and H3K27Ac was observed between mutant and wild type cell lines with epigenetic-targeted therapy, correlating with cell transcriptomes. CONCLUSIONS Histone H3 tail analysis reveals the effects of H3K27M mutation and bromodomain inhibition on the tumor epigenetic landscape, providing insight into mechanisms of tumorigenesis and therapy response. Further investigation of the utility of these signatures as biomarkers for diagnosis and monitoring treatment response are therefore underway.
- Published
- 2020
25. HGG-26. H3G34V MUTATION AFFECTS GENOMIC H3K36 METHYLATION IN PEDIATRIC GLIOMA
- Author
-
Rintaro Hashizume, Tina Huang, Andrea Piunti, Amanda Saratsis, Ali Shilatifard, Jin Qi, and Elizabeth T. Bartom
- Subjects
Genome instability ,Cancer Research ,Mutation ,Methylation ,Biology ,medicine.disease_cause ,medicine.disease ,Oncology ,Glioma ,Gene expression ,Cancer research ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,High Grade Glioma ,Carcinogenesis ,Allele frequency ,Gene - Abstract
BACKGROUND Histone H3.3 mutation (H3F3A) occurs in 50% of cortical pediatric high-grade gliomas. This mutation replaces glycine 34 with arginine or valine (G34R/V), impairing SETD2 activity (H3K36-specific trimethyltransferase), resulting in reduced H3K36me on H3G34V nucleosomes relative to wild-type. This contributes to genomic instability and drives distinct gene expressions associated with tumorigenesis. However, it is not known if this differential H3K36me3 enrichment is due to H3G34V mutant protein alone. Therefore, we set to elucidate the effect of H3G34V on genomic H3K36me3 enrichment in vitro. METHODS Doxycycline-inducible short hairpin RNA (shRNA) against H3F3A was delivered via lentivirus to established H3G34V mutant pediatric glioma cell line KNS42, and H3G34V introduced into H3.3 wild type normal human astrocytes (NHA). Transfections were confirmed by western blot, fluorescent imaging, and flow cytometry, with resulting H3.3WT and H3K36me3 expression determined by western blot. H3.3WT, H3K36me3, and H3G34V ChIP-Seq was performed to evaluate genomic enrichment. RESULTS Complete knockdown of H3G34V was achieved with DOX-induced shRNA, with no change in total H3.3, suggesting disproportionate allelic frequency of genes encoding H3.3 (H3F3A and H3F3B). Modest increase in H3K36me3 occurred after H3F3A-knockdown from KNS42, suggesting H3G34V alone impacts observed H3K36me3 levels. Distinct H3K36me3 genomic enrichment was observed with H3G34V knock-in. CONCLUSIONS We demonstrate that DOX-inducible knockdown of H3F3A in an H3G34V mutant pediatric glioma cells and H3G34V mutation transduction in wild-type astrocytes affects H3K36me3 expression. Further evaluation by ChIP-Seq analysis for restoration of wild-type genomic H3K36me3 enrichment patterns with H3G34V knockdown, and mutant H3K36me3 patterns with H3G34V transduction, is currently underway.
- Published
- 2020
26. CTIM-27. PHASE I/II STUDY OF CONTROLLED IL-12 AS IMMUNOTHERAPY FOR DIFFUSE INTRINSIC PONTINE GLIOMA (DIPG)
- Author
-
Laurence J.N. Cooper, Erin Walsh, John Miao, Arnold Gelb, Sabine Mueller, Rudy Allen, Amanda Saratsis, Nira Hadar, Stewart Goldman, Taylor Estupinan, Jill Buck, Nathan Demars, and Susan N. Chi
- Subjects
Cancer Research ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Clinical Trials: Immunologic ,Phase i ii ,Oncology ,medicine ,Interleukin 12 ,Cancer research ,Neurology (clinical) ,business ,Glioblastoma - Abstract
DIPG, which is the leading cause of pediatric brain cancer death with no effective treatment, has neither a highly immunosuppressive nor inflammatory tumor microenvironment (TME). Therefore, eliciting a pro-inflammatory TME may provide therapeutic benefit. We previously demonstrated in adults with recurrent glioblastoma that loco-regional delivery of interleukin 12 administered under the control of the proprietary transcriptional switch RheoSwitch Therapeutic Systemâ (RTSâ) delivered via a replication-incompetent adenovirus (“Controlled IL-12”) turned “cold” tumors “hot” for up to 5.8 months (Sci Transl Med. 2019;11(505)) and seemed to improve median overall survival as compared with historical controls (SNO 2020). A multicenter, phase I/II, open-label study (NCT03330197) is determining the safety and tolerability of Ad (2 x 1011 viral particles) administered by stereotactic intratumoral injection (Day 0) and 14 daily (Days 1 to 14) V doses (10 or 20 mg, body surface area adjusted). The first DIPG subject enrolled was in April 2020 with completion of the first cohort (arm 1, n=3) enrollment anticipated by September 2020. The first subject has tolerated treatment well with no SAEs during the evaluation period. Endogenous serum cytokines increased (including IFN-g 11.4 pg/mL, Day 3), consistent with V crossing the blood-brain barrier and activating the RTSâ switch to conditionally produce recombinant IL-12. Other biomarkers include plasma PK and circulating DNA. Follow-up is ongoing and enrollment is proceeding. Since development of effective immunotherapy for DIPG likely depends on eliciting a tumor-specific effector immune response, Controlled IL-12 is a promising immunotherapy candidate. The first DIPG subject shows encouraging data on safety, tolerability, serum cytokines and early signs consistent with a clinical response. After completion of dose-escalation, the study may be expanded up to 30 patients, which will be considered the phase II component of the study.
- Published
- 2020
27. Correction to: Comparative multidimensional molecular analyses of pediatric diffuse intrinsic pontine glioma reveals distinct molecular subtypes
- Author
-
Javad Nazarian, Melanie Weingart, Ping An, Brennan Harmon, Amanda Saratsis, Eric H. Raabe, Suresh N. Magge, Brian R. Rood, Tobey J. MacDonald, Sridevi Yadavilli, Roger J. Packer, Jordan Hall, Madhuri Kambhampati, Kendall Snyder, and Joseph M. Devaney
- Subjects
Cellular and Molecular Neuroscience ,Nuclear magnetic resonance ,Chemistry ,Neurology (clinical) ,Article ,Pathology and Forensic Medicine - Abstract
Diffuse Intrinsic Pontine Glioma (DIPG) is a highly morbid form of pediatric brainstem glioma. Here, we present the first comprehensive protein, mRNA, and methylation profiles of fresh frozen DIPG specimens (n=14), normal brain tissue (n=10), and other pediatric brain tumors (n=17). Protein profiling identified 2,305 unique proteins indicating distinct DIPG protein expression patterns compared to other pediatric brain tumors. Western blot and immunohistochemistry validated upregulation of Clusterin (CLU), Elongation Factor 2 (EF2), and Talin-1 (TLN1) in DIPGs studied. Comparisons to mRNA expression profiles generated from tumor and adjacent normal brain tissue indicated two DIPG subgroups, characterized by upregulation of Myc (N-Myc) or Hedgehog (Hh) signaling. We validated upregulation of PTCH, a membrane receptor in the Hh signaling pathway, in a subgroup of DIPG specimens. DNA methylation analysis indicated global hypomethylation of DIPG compared to adjacent normal tissue specimens, with differential methylation of 24 genes involved in Hh and Myc pathways, correlating with protein and mRNA expression patterns. Sequencing analysis showed c.83A>T mutations in the H3F3A or HIST1H3B gene in 77% of our DIPG cohort. Supervised analysis revealed a unique methylation pattern in mutated specimens compared to the wild type DIPG samples.
- Published
- 2020
28. PDTM-42. TARGETED INHIBITION OF BET BROMODOMAIN AND JMJD3 PROTEINS FOR THE TREATMENT OF DIFFUSE INTRINSIC PONTINE GLIOMA
- Author
-
Stewart Goldman, Kathryn E. Loughlin, Ali Zhang, Frank Eckerdt, Amanda Saratsis, Lihua Zou, Rintaro Hashizume, C. David James, Xingyao He, Gavin T. Blyth, Craig Horbinski, Patrick A. Ozark, Takahiro Sasaki, Hiroaki Katagi, Ali Shilatifard, and Rishi Lulla
- Subjects
Cancer Research ,Abstracts ,Oncology ,Chemistry ,Cancer research ,Neurology (clinical) ,Bromodomain - Abstract
Recent discovery of somatic histone gene mutations, resulting in replacement of lysine 27 by methionine (K27M) in the encoded histone H3.3 proteins, in diffuse intrinsic pontine glioma (DIPG) has dramatically improved our understanding of disease pathogenesis, and stimulated the development of novel therapeutic approaches targeting epigenetic regulators for disease treatment. K27M mutant DIPG shows a dramatic reduction in global methylation at K27 residues. We have shown that the JMJD3 demethylase inhibitor, GSKJ4, acted to restore K27 methylation in DIPG cells, while demonstrating potent anti-tumor activity, in vitro and in vivo. In addition to H3K27 methylation, H3K27 can also be acetylated (K27ac), which requires bromo- and extra-terminal domain (BET) protein activity. Increase level of H3K27 acetylation and bromodomain proteins in K27M-containing nucleosomes suggests that inhibitor of BET bromodomain protein 4 (BRD4), JQ1, could be useful for the treatment of K27M DIPG. Our aim is to investigate the hypothesis that the combined JQ1 + GSKJ4 have greater anti-tumor activity in vitro and in vivo than either monotherapy and reduce the likelihood of drug resistance. The level of H3K27 methylation and acetylation in DIPG cells with treated JQ1 + GSKJ4 were studied by western blotting. JQ1 + GSKJ4 increased H3K27 methylation and reduced K27acetylation in DIPG cells. MTS assay showed that combination treatment of JQ1 + GSKJ4 significantly increased growth inhibition, compared with either therapy alone. Colony formation assay showed that combination treatment of JQ1 + GSKJ4 significantly reduced the number of colonies. Boyden chamber assay showed that combination treatment of JQ1 + GSKJ4 significantly reduced cell invasion. Annexin V assay showed that combination treatment of JQ1 + GSKJ4 significantly promoted cell apoptosis. In vivo response to monotherapy and combination of JQ1 + GSKJ4 will be measured by bioluminescence imaging and animal survival studies in our human DIPG xenograft model.
- Published
- 2018
29. Detection of histone H3 K27M mutation and post-translational modifications in pediatric diffuse midline glioma via tissue immunohistochemistry informs diagnosis and clinical outcomes
- Author
-
Roxanna M. Garcia, Rishi Lulla, Charles David James, Tina Huang, Amanda Saratsis, Amir Behdad, Jin Qi, Craig Horbinski, Nitin R. Wadhwani, and Ali Shilatifard
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Mutation ,medicine.diagnostic_test ,business.industry ,medicine.disease_cause ,medicine.disease ,3. Good health ,03 medical and health sciences ,Histone H3 ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Glioma ,Biopsy ,medicine ,Transcriptional regulation ,Immunohistochemistry ,Epigenetics ,Carcinogenesis ,business ,030217 neurology & neurosurgery - Abstract
Pediatric diffuse midline glioma is a highly morbid glial neoplasm that may arise in the thalamus or brainstem (also known as diffuse intrinsic pontine glioma or DIPG). Because tumor anatomic location precludes surgical resection, diagnosis and treatment is based on MR imaging and analysis of biopsy specimens. Up to 80% of pediatric diffuse midline gliomas harbor a histone H3 mutation resulting in the replacement of lysine 27 with methionine (K27M) in genes encoding histone H3 variant H3.3 (H3F3A) or H3.1 (HIST1H3B). H3K27M mutant glioma responds more poorly to treatment and is associated with worse clinical outcome than wild-type tumors, so mutation detection is now diagnostic for a new clinical entity, diffuse midline glioma H3K27M mutant, as defined in the most recent WHO classification system. We previously reported patterns of histone H3 trimethylation (H3K27me3) and acetylation (H3K27Ac) associated with H3K27M mutation that impact transcription regulation and contribute to tumorigenesis. Given the clinical implications of the H3K27M mutation and these associated H3 post-translational modifications (PTMs), we set to determine whether they can be characterized via immunohistochemistry (IHC) in a cohort of pediatric glioma (n = 69) and normal brain tissue (n = 4) specimens. We observed 100% concordance between tissue IHC and molecular sequencing for detecting H3K27M mutation. In turn, H3K37M and H3K27me3 results, but not H3K27Ac staining patterns, were predictive of clinical outcomes. Our results demonstrate H3K27M and H3K27me3 staining of pediatric glioma tissue may be useful for diagnosis, stratification to epigenetic targeted therapies, and longitudinal monitoring of treatment response.
- Published
- 2018
30. DIPG-14. INTEGRATED DIFFUSE INTRINSIC PONTINE GLIOMA (DIPG) NEXT-GENERATION SEQUENCING REVEALS EPIGENETIC DYSREGULATION OF GENE EXPRESSION AND BROMODOMAIN INHIBITION AS A NOVEL THERAPEUTIC TARGET
- Author
-
C. David James, Rintaro Hashizume, Rishi Lulla, Elizaeth Bartom, Andrea Piunti, Tina Huang, Ali Shilatifard, Patrick A. Ozark, Amanda Saratsis, Stacy A. Marshall, and Jin Qi
- Subjects
Cancer Research ,Abstracts ,Oncology ,Gene expression ,Cancer research ,Neurology (clinical) ,Epigenetics ,Biology ,DNA sequencing ,Bromodomain - Abstract
INTRODUCTION: Diffuse intrinsic pontine glioma (DIPG) has the highest mortality of all pediatric solid tumors. Histone H3 mutation (H3K27M) occurs in 80%, altering chromatin structure and function. To characterize the effects of H3K27M as an oncogenic driver, we analyzed the transcriptome and epigenome in DIPG cell lines. METHODS: Transcriptomes (RNA-Seq) and genomic enrichment of Histone proteins (ChIP-Seq) were characterized in H3K27M DIPG (n=7), wild-type pediatric high-grade glioma (n=3), neural stem cells (n=1) and astrocyte cell lines (n=1). Reads were aligned (UCSC hg19, Tophat), normalized and quantified (HTSeq), and formatted \ for peak calling (SICER) and genomic enrichment mapping (Ensembl). Differential expression and enrichment patterns were identified (edgeR) then analyzed for biological relevance (TopGO, IPA). Epigenetic therapeutic targets were studied in vitro and in vivo. RESULTS: Unsupervised clustering revealed two DIPG subgroups, distinct from control and H3 wild-type lines. Increased EZH2 (p
- Published
- 2018
31. Methylation-dependent Tissue Factor Suppression Contributes to the Reduced Malignancy of IDH1-mutant Gliomas
- Author
-
Amanda Saratsis, Snezana Mirkov, Michael Drumm, C. David James, Kathleen McCortney, Craig Horbinski, Jann N. Sarkaria, Yuping D. Li, Dusten Unruh, Qazi F Haider, Jonathan B. Lamano, Rodrigo Javier, Denise M. Scholtens, and Brian Wray
- Subjects
0301 basic medicine ,Proteomics ,Transcriptional Activation ,Cancer Research ,congenital, hereditary, and neonatal diseases and abnormalities ,IDH1 ,Receptor tyrosine kinase ,Article ,Ectopic Gene Expression ,Epigenesis, Genetic ,Thromboplastin ,03 medical and health sciences ,Tissue factor ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Glioma ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Tumor Stem Cell Assay ,Gene knockdown ,biology ,DNA Methylation ,medicine.disease ,Isocitrate Dehydrogenase ,Demethylating agent ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,DNA methylation ,Mutation ,biology.protein ,Cancer research ,Neoplasm Grading ,Protein Binding - Abstract
Purpose: Gliomas with isocitrate dehydrogenase 1 mutations (IDH1mut) are less aggressive than IDH1 wild-type (IDH1wt) gliomas and have global genomic hypermethylation. Yet it is unclear how specific hypermethylation events contribute to the IDH1mut phenotype. Previously, we showed that the gene encoding the procoagulant tissue factor (TF), F3, is among the most hypermethylated and downregulated genes in IDH1mut gliomas, correlating with greatly reduced thrombosis in patients with IDH1mut glioma. Because TF also increases the aggressiveness of many cancers, the current study explored the contribution of TF suppression to the reduced malignancy of IDH1mut gliomas. Experimental Design: TF expression was manipulated in patient-derived IDH1mut and IDH1wt glioma cells, followed by evaluation of in vitro and in vivo behavior and analyses of cell signaling pathways. Results: A demethylating agent, decitabine, increased F3 transcription and TF-dependent coagulative activity in IDH1mut cells, but not in IDH1wt cells. TF induction enhanced the proliferation, invasion, and colony formation of IDH1mut cells, and increased the intracranial engraftment of IDH1mut GBM164 from 0% to 100% (P = 0.0001). Conversely, TF knockdown doubled the median survival of mice engrafted with IDH1wt/EGFRvIIIamp GBM6, and caused complete regression of IDH1wt/EGFRamp GBM12 (P = 0.001). In vitro and in vivo effects were linked to activation of receptor tyrosine kinases (RTK) by TF through a Src-dependent intracellular pathway, even when extracellular RTK stimulation was blocked. TF stimulated invasion predominately through upregulation of β-catenin. Conclusions: These data show that TF suppression is a component of IDH1mut glioma behavior, and that it may therefore be an attractive target against IDH1wt gliomas.
- Published
- 2018
32. The emerging role of NG2 in pediatric diffuse intrinsic pontine glioma
- Author
-
Jyoti K. Jaiswal, Javad Nazarian, Joseph Scafidi, Tobey J. MacDonald, Madhuri Kambhampati, Kari Elise T. Codispoti, Santi Mariarita, Amanda Saratsis, Oren J. Becher, Roger J. Packer, Sridevi Yadavilli, Rebecca L. Hiner, and Suresh N. Magge
- Subjects
Adolescent ,Brain Stem Neoplasm ,Brain tumor ,Biology ,OLIG2 ,Mice ,NG2 ,Glioma ,medicine ,Animals ,Brain Stem Neoplasms ,Humans ,Antigens ,Child ,histone 3 ,Gene Expression Profiling ,Cancer ,PDGF ,medicine.disease ,Primary tumor ,nervous system ,Oncology ,Mutation ,Behavioral medicine ,DIPG ,Cancer research ,Proteoglycans ,Research Paper ,Biomedical sciences - Abstract
// Sridevi Yadavilli 1 , Joseph Scafidi 2 , Oren J. Becher 3 , Amanda M. Saratsis 4 , Rebecca L. Hiner 5 , Madhuri Kambhampati 1 , Santi Mariarita 6 , Tobey J. MacDonald 7 , Kari-Elise Codispoti 8 , Suresh N. Magge 9 , Jyoti K. Jaiswal 1,10 , Roger J. Packer 11 and Javad Nazarian 1,10 1 Research Center for Genetic Medicine, Children’s National Health System, Washington, DC, USA 2 Department of Neurology and Center for Neuroscience Research, Children’s National Health System, Washington, DC, USA 3 Department of Pediatrics and Pathology, Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA 4 Division of Neurosurgery, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA 5 Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, NY, USA 6 Department of Pathology and Lab Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA 7 Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA 8 Department of Pathology, Children’s National Health System, Washington, DC, USA 9 Division of Neurosurgery, Children’s National Health System, Washington, DC, USA 10 Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA 11 Brain Tumor Institute, Center for Neuroscience and Behavioral Medicine, Children’s National Health System, Washington, DC, USA Correspondence to: Javad Nazarian, email: // Keywords : DIPG, NG2, PDGF, histone 3, glioma Received : March 04, 2015 Accepted : March 11, 2015 Published : March 30, 2015 Abstract Diffuse intrinsic pontine gliomas (DIPGs) have a dismal prognosis and are poorly understood brain cancers. Receptor tyrosine kinases stabilized by neuron-glial antigen 2 (NG2) protein are known to induce gliomagenesis. Here, we investigated NG2 expression in a cohort of DIPG specimens (n= 50) . We demonstrate NG2 expression in the majority of DIPG specimens tested and determine that tumors harboring histone 3.3 mutation express the highest NG2 levels. We further demonstrate that microRNA 129-2 (miR129-2) is downregulated and hypermethylated in human DIPGs, resulting in the increased expression of NG2. Treatment with 5-Azacytidine, a methyltransferase inhibitor, results in NG2 downregulation in DIPG primary tumor cells in vitro . NG2 expression is altered (symmetric segregation) in mitotic human DIPG and mouse tumor cells. These mitotic cells co-express oligodendrocyte (Olig2) and astrocyte (glial fibrillary acidic protein, GFAP) markers, indicating lack of terminal differentiation. NG2 knockdown retards cellular migration in vitro, while NG2 expressing neurospheres are highly tumorigenic in vivo, resulting in rapid growth of pontine tumors. NG2 expression is targetable in vivo using miR129-2 indicating a potential avenue for therapeutic interventions. This data implicates NG2 as a molecule of interest in DIPGs especially those with H3.3 mutation.
- Published
- 2015
33. Putting Residents in the Office: An Effective Method to Teach the Systems-Based Practice Competency
- Author
-
Vinod Narra, A. Alfred Chahine, Marisa E. Pulcrano, Amanda Saratsis, Jamie Divine-Cadavid, and Stephen R. T. Evans
- Subjects
Adult ,Male ,Office Management ,education ,Graduate medical education ,Pilot Projects ,Education ,Hospitals, University ,Appointments and Schedules ,Ambulatory care ,Humans ,Outpatient clinic ,Medicine ,Effective method ,Program Development ,Curriculum ,Accreditation ,Medical education ,business.industry ,Core competency ,Internship and Residency ,Problem-Based Learning ,Competency-Based Education ,Ambulatory Surgical Procedures ,Education, Medical, Graduate ,General Surgery ,Test score ,District of Columbia ,Female ,Surgery ,Clinical Competence ,business ,Program Evaluation - Abstract
Objectives Systems-based practice (SBP) was 1 of 6 core competencies established by the Accreditation Council for Graduate Medical Education and has proven to be one of the most difficult to effectively implement. This pilot study presents an immersion workshop as an effective tool to teach the SBP competency in a way that could easily be integrated into a residency curriculum. Design In 2006, 16 surgical residents rotated through 3 stations for 30 minutes each: coding and billing, scheduling operations and return appointments, and patient check-in. Participants were administered a pretest and posttest questionnaire evaluating their knowledge of SBP, and were asked to evaluate the workshop. Setting Outpatient clinic at MedStar Georgetown University Hospital, Washington, DC. Participants Residents in the general surgery residency training program at MedStar Georgetown University Hospital. Results Most residents (62.5%) improved their score after the workshop, whereas 31.25% showed no change and 6.25% demonstrated a decrease in score. Overall within their training levels, all groups demonstrated an increase in mean test score. Postgraduate year-2 residents demonstrated the greatest change in mean score (20%), whereas postgraduate year-4 residents demonstrated the smallest change in mean score (3.3%). Conclusions An immersion workshop where general surgery residents gained direct exposure to SBP concepts in situ was an effective and practical method of integrating this core competency into the residency curriculum. Such a workshop could complement more formal didactic teaching and be easily incorporated into the curriculum. For example, this workshop could be integrated into the ambulatory care requirement that each resident must fulfill as part of their clinical training.
- Published
- 2015
34. PDTM-44. RADIATION DNA DAMAGE REPAIR INHIBITION BY GSK-J4 INDUCED CHROMATIN COMPACTION IN DIPG
- Author
-
Nundia Louis, Xingyao He, Andrea Piunti, Marc Morgan, Dusten Unruh, Amanda Saratsis, Rishi Lulla, Jason R Fangusaro, Craig Horbinski, Stewart Goldman, C David James, Ali Shilatifard, and Rintaro Hashizume
- Subjects
Cancer Research ,Abstracts ,Oncology ,Neurology (clinical) - Published
- 2017
35. Immunohistochemical analysis of H3K27me3 demonstrates global reduction in group-A childhood posterior fossa ependymoma and is a powerful predictor of outcome
- Author
-
Shayna Zelcer, Cynthia Hawkins, Jill Bayliss, Nada Jabado, Alexander R. Judkins, Sarah J. Curry, Christopher Dunham, Jonathan Clark, Melike Pekmezci, Lukas Chavez, Pooja Panwalkar, Chan Chung, Donna L. Johnston, Stephen Yip, Juliette Hukin, Lucie Lafay-Cousin, Adam Banda, Marcel Kool, Matija Snuderl, Mariarita Santi, Fausto J. Rodriguez, P. Daniel McNeely, Ashley Margol, Vijay Ramaswamy, Craig Horbinski, Matthias A. Karajannis, Andrey Korshunov, Sriram Venneti, Stefan M. Pfister, Amanda Saratsis, Katrin Scheinemann, David D. Eisenstat, Anne Sophie Carret, Yilun Sun, M. Silva, Michael D. Taylor, Fusheng Yang, Debra Hawes, Matthew J. Schipper, and Beverly Wilson
- Subjects
Ependymoma ,Male ,Childhood ependymoma ,Pathology ,Jumonji Domain-Containing Histone Demethylases ,H3K27me3 ,Posterior fossa ,Infratentorial Neoplasms ,Pediatrics ,Group A ,0302 clinical medicine ,Registries ,Child ,Cancer ,Pediatric ,Prognosis ,Survival Rate ,Child, Preschool ,030220 oncology & carcinogenesis ,Immunohistochemistry ,Female ,Epigenetics ,Molecular subgrouping ,Poor prognosis ,medicine.medical_specialty ,Pediatric Cancer ,Clinical Trials and Supportive Activities ,Clinical Sciences ,macromolecular substances ,Biology ,Disease-Free Survival ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Rare Diseases ,Clinical Research ,medicine ,Genetics ,Humans ,Preschool ,Survival rate ,Neurology & Neurosurgery ,Neurosciences ,Infant ,medicine.disease ,Brain Disorders ,Brain Cancer ,Neurology (clinical) ,030217 neurology & neurosurgery ,Immunostaining - Abstract
Posterior fossa ependymomas (EPN_PF) in children comprise two morphologically identical, but biologically distinct tumor entities. Group-A (EPN_PFA) tumors have a poor prognosis and require intensive therapy. In contrast, group-B tumors (EPN_PFB) exhibit excellent prognosis and the current consensus opinion recommends future clinical trials to test the possibility of treatment de-escalation in these patients. Therefore, distinguishing these two tumor subtypes is critical. EPN_PFA and EPN_PFB can be distinguished based on DNA methylation signatures, but these assays are not routinely available. We have previously shown that a subset of poorly prognostic childhood EPN_PF exhibits global reduction in H3K27me3. Therefore, we set out to determine whether a simple immunohistochemical assay for H3K27me3 could be used to segregate EPN_PFA from EPN_PFB tumors. We assembled a cohort of 230 childhood ependymomas and H3K27me3 immunohistochemistry was assessed as positive or negative in a blinded manner. H3K27me3 staining results were compared with DNA methylation-based subgroup information available in 112 samples [EPN_PFA (n=72) and EPN_PFB tumors (n=40)]. H3K27me3 staining was globally reduced in EPN_PFA tumors and immunohistochemistry showed 99% sensitivity and 100% specificity in segregating EPN_PFA from EPN_PFB tumors. Moreover, H3K27me3 immunostaining was sufficient to delineate patients with worse prognosis in two independent, non-overlapping cohorts (n=133 and n=97). In conclusion, immunohistochemical evaluation of H3K27me3 global reduction is an economic, easily available and readily adaptable method for defining high-risk EPN_PFA from low-risk posterior fossa EPN_PFB tumors to inform prognosis and to enable the design of future clinical trials.
- Published
- 2017
36. PDTM-49. GLOBAL REDUCTION IN H3K27me3, SIMILAR TO H3K27M MUTANT GLIOMAS, IS A MOLECULAR SURROGATE FOR PEDIATRIC POSTERIOR FOSSA- GROUP A EPENDYMOMAS
- Author
-
Amanda Saratsis, Sriram Vennneti, Debra Hawes, Ashley Margol, Stefan M. Pfister, Andrey Korshunov, Sarah J. Curry, Juliette Hukin, Matthias A. Karajannis, Pooja Panwalkar, Chan Chung, Jessica Clark, Yilun Sun, Ramaswamy, Fausto J. Rodriguez, Craig Horbinski, Michael D. Taylor, Marcel Kool, Cynthia Hawkins, Mariarita Santi, Christopher Dunham, Alexander R. Judkins, Stephen Yip, Adam Banda, Jill Bayliss, Lukas Chavez, Matija Snuderl, Fusheng Yang, Matthew J. Schipper, and Melike Pekmezci
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Mutant ,Posterior fossa ,macromolecular substances ,Group A ,Abstracts ,Text mining ,Oncology ,medicine ,Neurology (clinical) ,business ,Reduction (orthopedic surgery) - Abstract
Ependymomas in children occur most commonly in the posterior fossa of the brain and cause significant morbidity and mortality. The pathogenesis of these tumors remains obscure as recent whole genome and whole exome sequencings have not yielded recurrent genetic alterations. These tumors are classified into PFA and PFB groups based on abnormal CpG island methylation suggesting that epigenetic alterations may be a significant driver. To gain insights into the epigenetics of childhood ependymomas, we performed mass spectroscopy for histone modifications to discover that H3K27me3 was globally reduced in PFA tumors, similar to that observed in H3K27M diffuse intrinsic pontine gliomas (DIPGs). Despite global reduction in H3K27me3, ChIP-sequencing for H3K27me3 revealed genomic enrichment at several loci important for neurodevelopment. Comparison of DNA methylation and genome-wide H3K27me3 enrichment from both H3K27M mutant DIPGs and PFAs showed many similarities and pointed to factors that regulate radial glial (neuronal stem cells) development. Moreover, radial glia in the developing human posterior fossa were negative for H3K27me3. To assess the clinical utility of this finding, H3K27me3 immunohistochemistry was assessed as positive or negative in a blinded manner in a cohort (n=112) of previously defined childhood PFA (n=72) and PFB tumors (n=40). Next, H3K27me3 immunohistochemistry status in two independent, non-overlapping cohorts of childhood EPN (n=133 and n=98) was determined and risk stratification was assessed retrospectively using univariate and multivariate Cox proportional hazards analyses. H3K27me3 immunohistochemistry showed 99% sensitivity and 100% specificity in delineating PFA from PFB tumors and was sufficient to delineate patients with worse prognosis (P
- Published
- 2017
37. PDTM-28. TARGETED INHIBITION OF EZH2 AND BET BROMODOMAIN PROTEINS FOR THE TREATMENT OF DIFFUSE INTRINSIC PONTINE GLIOMAS
- Author
-
Ali Zhang, Nundia Louis, Xingyao He, Rintaro Hashizume, Andrea Piunti, Rishi Lulla, Emily J. Rendleman, Marc A. Morgan, Yoh Hei Takahashi, Elizabeth T. Bartom, Nebiyu Abshiru, Alexander V. Misharin, Craig Horbinski, Stacy A. Marshall, Amanda Saratsis, Ali Shilatifard, Neil L. Kelleher, and C. David James
- Subjects
0301 basic medicine ,Cancer Research ,biology ,Chemistry ,EZH2 ,macromolecular substances ,medicine.disease ,Pons ,Bromodomain ,03 medical and health sciences ,Histone H3 ,Abstracts ,030104 developmental biology ,Histone ,Oncology ,Tumor progression ,Glioma ,Neuron differentiation ,biology.protein ,medicine ,Cancer research ,Neurology (clinical) - Abstract
Recent discovery of somatic histone gene mutations, resulting in replacement of lysine 27 by methionine (K27M) in the encoded histone H3.3 proteins, in diffuse intrinsic pontine glioma (DIPG) has dramatically improved our understanding of disease pathogenesis, and stimulated the development of novel therapeutic approaches targeting epigenetic regulators for disease treatment. K27M mutant DIPG shows a dramatic reduction in global methylation at K27 residues on all 16 H3 proteins. This reduction in H3K27 methylation is believed to modify cellular gene expression in a way that favors tumor development. We have shown that inhibition of the H3K27 demethylase JMJD3 acts to restore K27 methylation in DIPG cells, while demonstrating potent anti-tumor activity, in vitro and in vivo. In addition to H3K27 methylation, H3K27 can also be acetylated (K27ac), which requires bromo- and extra-terminal domain (BET) protein activity. We have recently identified H3K27M-K27ac nucleosomes co-localize with BET bromodomain proteins at actively transcribed genes, whereas a polycomb repressive complex 2 (PRC2) is excluded from these regions, demonstrating that H3K27M and PRC2 occupy distinct chromatin regions in DIPG cells. Despite a major loss of H3K27 methylation, PRC2 activity is still detected in H3K27M DIPG cells, and the residual PRC2 activity is required to maintain DIPG proliferative potential by repressing neuronal differentiation and function. Small molecule inhibitor of EZH2 inhibited cell growth through the upregulation of gene that are normally repressed by PRC2 in DIPG. We also tested the anti-tumor activity of BET bromodomain containing protein 4 (BRD4) inhibitor, JQ1, and EZH2 inhibitor, GSK126, in our human DIPG xenograft model, and JQ1 and GSK126 treatment results in a significant delay of tumor progression and prolonged animal survival. Our findings suggest EZH2 inhibition could be a potential combinatorial strategy with BRD4 inhibition in treating K27M DIPG, that could counteract the development of resistance to single-agent epigenetic therapy.
- Published
- 2017
38. DIPG-25. IN VITRO AND IN VIVO ANALYSIS OF TENASCIN-C EXPRESSION IN PEDIATRIC BRAINSTEM GLIOMA
- Author
-
Tadanori Tomita, C. David James, Tina Huang, Jin Qi, Rintaro Hashizume, and Amanda Saratsis
- Subjects
Cancer Research ,biology ,business.industry ,Tenascin C ,In vivo analysis ,medicine.disease ,musculoskeletal system ,Pediatric Brainstem Glioma ,In vitro ,Abstracts ,Oncology ,Glioma ,Gene expression ,Cancer research ,biology.protein ,Medicine ,Immunohistochemistry ,Neurology (clinical) ,business ,Protein overexpression - Abstract
INTRODUCTION: Diffuse intrinsic pontine glioma (DIPG) is a highly morbid pediatric brainstem tumor. Tenascin-C (TNC) is an extracellular matrix protein associated with NOTCH pathway activation, and is expressed during normal brain development by oligodendroglial cells (OPCs), the purported DIPG cell of origin. We previously reported TNC overexpression in DIPG tissue and CSF, associated with Histone H3K27M mutation. We therefore investigated the pattern and effect of TNC expression using pediatric glioma primary cell lines. METHODS: To determine the molecular effects of altered TNC expression in DIPG in vitro, we evaluated endogenous TNC expression in DIPG cell lines (n=3) and compared to TNC suppression and over-expression via vector-mediated shRNA knockdown and cDNA amplification, respectively. Effects of genetic manipulations were confirmed by western blot and qPCR. Cell proliferation, migration and adhesion were compared, as well as patterns of gene expression (RNA-Seq) across cell lines. Luciferase-expressing DIPG cells modified with TNC shRNA were used to create mouse intracranial xenografts. Animal survival and tumor growth was compared between groups, and TNC expression confirmed by immunohistochemistry. RESULTS: shRNA knockdown achieved up to 80% reduction in TNC expression, inhibiting cell proliferation and migration on functional analysis compared to control DIPG lines. Distinct pathways of gene expression favoring dedifferentiation and stem-like state correlated with higher TNC expression. Anaylsis cDNA and in vivo studies is currently underway. CONCLUSIONS: Here, we report the pattern and effect of modulated TNC expression in pediatric brainstem glioma (DIPG). Our findings suggest TNC may play a role in cell proliferation, migration, and maintaining stem-ness, possibly contributing to brainstem gliomagenesis. Further in vitro and in vivo studies exploring the mechanism of overexpression and effects of targeting TNC expression in DIPG are planned.
- Published
- 2017
39. DIPG-24. EPIGENETIC ANALYSIS OF FORMALIN-FIXED PARAFFIN-EMBEDDED PEDIATRIC GLIOMA TISSUE
- Author
-
Tina Huang, Ali Shilatifard, Andrea Piunti, C. David James, Rintaro Hashizume, Jin Qi, Elizabeth T. Bartom, Tadanori Tomita, and Amanda Saratsis
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Formalin fixed paraffin embedded ,business.industry ,Epigenetic Analysis ,Brain stem glioma ,Abstracts ,Oncology ,Formalin-fixed paraffin-embedded tissue specimen ,Pediatric glioma ,Medicine ,Neurology (clinical) ,business - Abstract
BACKGROUND: Diffuse midline gliomas are uniquely characterized by missense mutations in Histone H3 isoforms H3.3 and H3.1, associated with distinct gene, protein and methylation profiles and poorer clinical outcomes. As a result, mutant H3 protein is of great interest as a potential driver of pediatric gliomagenesis. Importantly, mutant tumors harbor distinct patterns of post-translational modification (PTM) at key regulatory H3 amino acid residues affecting chromatin structure and function, and control of gene transcription. Chromatin immunoprecipitation sequencing (ChIP-Seq) is a powerful tool to evaluate genomic effects of histone PTMs. However, ChIP-Seq of diffuse midline glioma is limited due to the scarcity of fresh tumor tissue available for analysis, while primary tumor cell lines may not accurately reflect the tumor epigenome. METHODS: Our group recently developed a protocol that overcomes heavy DNA crosslinking and fragmentation characteristic of formalin-fixed, paraffin-embedded (FFPE) tissue to achieve successful chromatin extraction and sequencing. Using this approach, we performed the first successful extraction of chromatins from FFPE pediatric brain tumor tissue, observing genomic enrichment of H3K4me3 and H3K27ac via ChIP-Seq in a Juvenille Pilocytic Astrocytoma (JPA). We then applied this novel approach to rare diffuse midline glioma specimens. RESULTS: We applied our novel ChIP-Seq protocol to FFPE cell pellets from pediatric brainstem glioma (H3K27M) and supratentorial GBM (H3K27 wild-type) primary cell lines. Genomic enrichment of H3, H3K27ac and H3K27me3 is compared to corresponding fresh cells, FFPE mouse xenograft and subcutaneous tumor tissue derived from the same primary cell lines. ChIP-Seq of rare FFPE brainstem glioma specimens is now underway. CONCLUSIONS: Consistency of ChIP-Seq results from fresh and archival samples demonstrates the technical feasibility and biological relevance of FFPE tissue ChIP-Seq analysis. Our results suggest FFPE ChIP-Seq may serve as an alternative to fresh tumor analysis for elucidating genomic H3 deposition and tumor-specific patterns of H3 PTMs.
- Published
- 2017
40. DIPG-28. RADIATION DNA DAMAGE REPAIR INHIBITION BY GSK-J4 INDUCED CHROMATIN COMPACTION IN DIPG
- Author
-
Nundia Louis, Xingyao He, Andrea Plunti, Amanda Saratsis, Rishi Lulla, Jason Fangusaro, Craig Horbinski, Stewart Goldman, David James, Ali Shilatifard, and Rintaro Hashizume
- Subjects
Cancer Research ,Abstracts ,Oncology ,Neurology (clinical) - Abstract
INTRODUCTION: Focal radiation therapy has long been and still remains the only treatment option for diffuse intrinsic pontine glioma (DIPG). However, all children who suffer from this inoperable and uniformly fatal cancer show evidence of disease progression within months of completing radiation therapy, especially those who harbor the H3.3K27M mutation. Since chemotherapy does not provide any significant outcome improvement, it is crucial to find a suitable radiosensitizer. Our research has shown that the JMJD3 demethylase inhibitor, GSK-J4, not only exerts potent anti-tumor activity on H3K27M mutant DIPG cells, but also restores methylation while decreasing genes that encode proteins involved in DNA double-strand break (DSB) repair. Our aim is to investigate the hypothesis that GSK-J4 may inhibit radiation-induced DNA repair via chromatin modification, making it a potential radiosensitizing agent. METHODS: We evaluated DNA damage repair via quantitative-PCR (q-PCR), immunocytochemistry of DSB markers γH2AX and 53BP1, and DNA repair assay. MNase digestion was used to analyze nucleosome assembly in cellular chromatin. We are currently conducting western blotting, DNA repair assay, FAIRE-seq and clonogenic survival assays to assess DIPG response to radiation therapy (RT) in combination with GSK-J4. In vivo response to radiation monotherapy and combination of RT + GSK-J4 will be measured by bioluminescence imaging and animal survival studies. RESULTS: qPCR results showed that GSK-J4 significantly reduces DNA DSB repair genes such as PARP1, PARP2, SMARCB1 and BRCA1, in irradiated DIPG cells. Immunocytochemistry results support that GSK- J4 sustains high levels of γH2AX and 53BP1 in irradiated DIPG cells, thereby inhibiting DNA DSB repair. MNase assay indicated that GSK-J4 may cause chromatin compaction which prevents DNA DSB repair. Results from the clonogenic assays, to assess long term effect of GSK-J4 on radiated cells, as well as western blotting, FAIRE-seq and in vivo studies will be reported at the meeting.
- Published
- 2017
41. Detection of Histone H3 mutations in cerebrospinal fluid-derived tumor DNA from children with diffuse midline glioma
- Author
-
Amanda Saratsis, Tina Huang, Ali Shilatifard, Jin Qi, Tadanori Tomita, Andrea Piunti, Rishi Lulla, Craig Horbinski, and C. David James
- Subjects
Pathology ,medicine.medical_specialty ,DNA Mutational Analysis ,Biology ,Gene mutation ,medicine.disease_cause ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,Histones ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Histone H3 ,symbols.namesake ,0302 clinical medicine ,Diffuse intrinsic pontine glioma (DIPG) ,H3K27M ,Glioma ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Humans ,Liquid biopsy ,lcsh:Neurology. Diseases of the nervous system ,Sanger sequencing ,Mutation ,Brain Neoplasms ,Research ,Brain ,Diffuse midline glioma ,DNA, Neoplasm ,medicine.disease ,Immunohistochemistry ,3. Good health ,Histone ,Cerebrospinal fluid ,030220 oncology & carcinogenesis ,biology.protein ,symbols ,Feasibility Studies ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
Diffuse midline gliomas (including diffuse intrinsic pontine glioma, DIPG) are highly morbid glial neoplasms of the thalamus or brainstem that typically arise in young children and are not surgically resectable. These tumors are characterized by a high rate of histone H3 mutation, resulting in replacement of lysine 27 with methionine (K27M) in genes encoding H3 variants H3.3 (H3F3A) and H3.1 (HIST1H3B). Detection of these gain-of-function mutations has clinical utility, as they are associated with distinct tumor biology and clinical outcomes. Given the paucity of tumor tissue available for molecular analysis and relative morbidity of midline tumor biopsy, CSF-derived tumor DNA from patients with diffuse midline glioma may serve as a viable alternative for clinical detection of histone H3 mutation. We demonstrate the feasibility of two strategies to detect H3 mutations in CSF-derived tumor DNA from children with brain tumors (n = 11) via either targeted Sanger sequencing of H3F3A and HIST1H3B, or H3F3A c.83 A > T detection via nested PCR with mutation-specific primers. Of the six CSF specimens from children with diffuse midline glioma in our cohort, tumor DNA sufficient in quantity and quality for analysis was isolated from five (83%), with H3.3K27M detected in four (66.7%). In addition, H3.3G34V was identified in tumor DNA from a patient with supratentorial glioblastoma. Test sensitivity (87.5%) and specificity (100%) was validated via immunohistochemical staining and Sanger sequencing in available matched tumor tissue specimens (n = 8). Our results indicate that histone H3 gene mutation is detectable in CSF-derived tumor DNA from children with brain tumors, including diffuse midline glioma, and suggest the feasibility of “liquid biopsy” in lieu of, or to complement, tissue diagnosis, which may prove valuable for stratification to targeted therapies and monitoring treatment response. Electronic supplementary material The online version of this article (doi:10.1186/s40478-017-0436-6) contains supplementary material, which is available to authorized users.
- Published
- 2017
42. Therapeutic targeting of polycomb and BET bromodomain proteins in diffuse intrinsic pontine gliomas
- Author
-
Ali Shilatifard, Emily J. Rendleman, Quanhong Ma, Neil L. Kelleher, Amanda Saratsis, Elizabeth T. Bartom, Rintaro Hashizume, C. David James, Ashley R. Woodfin, Alexander V. Misharin, Nebiyu Abshiru, Rishi Lulla, Craig Horbinski, Stacy A. Marshall, Yoh Hei Takahashi, Marc A. Morgan, and Andrea Piunti
- Subjects
0301 basic medicine ,Epigenomics ,Neurogenesis ,macromolecular substances ,Methylation ,General Biochemistry, Genetics and Molecular Biology ,Article ,Histones ,03 medical and health sciences ,Histone H3 ,Mice ,Cell Line, Tumor ,Animals ,Brain Stem Neoplasms ,Humans ,Molecular Targeted Therapy ,Cell Proliferation ,biology ,Polycomb Repressive Complex 2 ,RNA-Binding Proteins ,Acetylation ,General Medicine ,Epigenome ,Azepines ,Glioma ,Triazoles ,Molecular biology ,Xenograft Model Antitumor Assays ,Chromatin ,Bromodomain ,Nucleosomes ,Gene Expression Regulation, Neoplastic ,Histone Code ,Protein Transport ,030104 developmental biology ,Histone ,Tumor progression ,Mutation ,biology.protein ,Cancer research ,PRC2 - Abstract
Diffuse intrinsic pontine glioma (DIPG) is a highly aggressive pediatric brainstem tumor characterized by rapid and uniform patient demise. A heterozygous point mutation of histone H3 occurs in more than 80% of these tumors and results in a lysine-to-methionine substitution (H3K27M). Expression of this histone mutant is accompanied by a reduction in the levels of polycomb repressive complex 2 (PRC2)-mediated H3K27 trimethylation (H3K27me3), and this is hypothesized to be a driving event of DIPG oncogenesis. Despite a major loss of H3K27me3, PRC2 activity is still detected in DIPG cells positive for H3K27M. To investigate the functional roles of H3K27M and PRC2 in DIPG pathogenesis, we profiled the epigenome of H3K27M-mutant DIPG cells and found that H3K27M associates with increased H3K27 acetylation (H3K27ac). In accordance with previous biochemical data, the majority of the heterotypic H3K27M-K27ac nucleosomes colocalize with bromodomain proteins at the loci of actively transcribed genes, whereas PRC2 is excluded from these regions; this suggests that H3K27M does not sequester PRC2 on chromatin. Residual PRC2 activity is required to maintain DIPG proliferative potential, by repressing neuronal differentiation and function. Finally, to examine the therapeutic potential of blocking the recruitment of bromodomain proteins by heterotypic H3K27M-K27ac nucleosomes in DIPG cells, we performed treatments in vivo with BET bromodomain inhibitors and demonstrate that they efficiently inhibit tumor progression, thus identifying this class of compounds as potential therapeutics in DIPG.
- Published
- 2017
43. PDTM-39. CRACKING THE HISTONE CODE: REVEALING THERAPEUTIC EPIGENETIC TARGETS THROUGH HISTONE H3 TAIL ANALYSIS OF DIFFUSE INTRINSIC PONTINE GLIOMA
- Author
-
Daphne Li, Tina Huang, Jin Qi, Amanda Saratsis, Jeannie M. Camarillo, Hannah Weiss, Paul M. Thomas, and Javad Nazarian
- Subjects
Cancer Research ,Histone H3 ,Oncology ,Pediatric Tumors ,Chemistry ,Histone code ,Neurology (clinical) ,Epigenetics ,Cell biology - Abstract
INTRODUCTION Diffuse intrinsic pontine glioma (DIPG) is a highly morbid pediatric cancer. Up to 80% harbor a Histone H3K27M mutation, which alters wild type Histone H3 protein post-translational modifications (PTMs) and genomic enrichment patterns to impact chromatin structure and transcription regulation. We previously identified tumorigenic patterns of H3K27Ac/bromodomain co-enrichment in DIPG, and demonstrated pre-clinical efficacy of bromodomain inhibition (JQ1). Here, we employ a novel proteomics platform, developed at our institution, to further elucidate the impact of H3K7M mutation on glioma histone codes and response to bromodomain inhibition. METHODS Epiproteomic analysis was performed on pediatric glioma cell lines (H3K27 WT n=2, H3K27M n=2) to characterize 95 distinct Histone H3.3 and H3.1 N-terminal tail modification states. Cells were treated with JQ1 or DMSO, and collected at 0h, 24h, 48h. Histones were extracted from isolated nuclei, immunopurified, and analyzed by LC-MS/MS. Results were integrated with RNA-Seq and ChIP Seq results (H3K27M, H3K27Ac, H3K27me3, H3K4me1, H3K4me3) from the same DIPG cell lines. Pediatric glioma tissues (H3K27M WT n=3, H3K27M n= 9) were similarly analyzed to validate cell line results. RESULTS Cell PTM profiles cluster by H3 mutation status on unsupervised analysis. Relative H3 PTM abundance were compared across cell lines by tumor location, H3 mutation status, and in response to treatment. Significant differential genomic enrichment H3K27M and H3.3 WT proteins, H3K27Me3 and H3K27Ac were observed between mutant and wild type cell lines with epigenetic-targeted therapy, correlating with cell transcriptomes. CONCLUSIONS Histone H3 tail epiproteomic analysis reveals DIPG histone codes in situ, revealing the effects of bromodomain inhibition on the tumor epigenetic landscape and providing new insight to the mechanism of tumor formation and therapy response. Further investigation of the utility of these signatures as biomarkers for diagnosis and longitudinal monitoring of treatment response are therefore currently underway.
- Published
- 2019
44. PDTM-42. CROSS-PLATFORM PROFILING OF ctDNA USING ddPCR: STANDARDIZATION OF THE LIQUID BIOPSY FOR PEDIATRIC DIFFUSE MIDLINE GLIOMA
- Author
-
Rishi Lulla, Daphne Li, Stefanie Stallard, Carl Koschmann, Javad Nazarian, Eshini Panditharatna, Tina Huang, Amanda Saratsis, and Erin R. Bonner
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Oncology ,Pediatric Tumors ,business.industry ,Glioma ,medicine ,Profiling (information science) ,Neurology (clinical) ,Liquid biopsy ,medicine.disease ,business - Abstract
INTRODUCTION Pediatric diffuse midline glioma (DMG) with histone H3 mutations (80%), are highly morbid tumors with poor response to therapy. We previously detected H3 mutations in circulating tumor DNA (ctDNA) from CSF derived from children with DMG and high grade glioma. Here, we describe a high-throughput, sensitive and specific approach for H3 mutation detection and quantification in plasma and CSF specimens, validated across multiple centers. METHODS DNA extracted from tissue-validated H3.3K27M specimens (4 tumor, 5 CSF, 4 plasma) and pediatric glioma cell lines (1 wild-type, 1 H3.3K27M) were used to standardize ddPCR workflows. ctDNA extracted from 500uL of CSF or plasma was pre-amplified using sequence-specific primers, then analyzed on RainDance and BioRad ddPCR systems using two sets of custom primers and fluorescent LNA probes (Assays A and B). H3 mutation allelic frequency (MAF) was determined across specimens, and validated through parallel analysis of additional matched H3.3K27M tumor tissue, CSF and plasma specimens (n=3). RESULTS ctDNA processing and detection was standardized across three institutions and two platforms with sensitive, specific and reproducible H3 mutation detection. H3K27M mutations were detected in as little as 0.3-2pg input DNA on both platforms, with comparable MAFs across instruments and assays. RainDance yielded greater overall positive droplet number detection (increased sensitivity). Differences in optimal sample input volume between platforms were circumvented by vacuum concentration or dilution to maintain equal input DNA. Assay A yielded superior specificity so was used for subsequent matched specimen analysis. Mutation detection in plasma, as in previous studies, remained challenging due to low [ctDNA], while CSF analysis yielded reliable results across assays and platforms. CONCLUSIONS We demonstrate the utility of liquid biopsy for identifying H3K27M mutations in plasma and CSF via ddPCR with low starting [DNA], representing a rapid, minimally invasive method for diagnosis and therapeutic monitoring of DMG.
- Published
- 2019
45. Correction: Detection of histone H3 K27M mutation and post-translational modifications in pediatric diffuse midline glioma via tissue immunohistochemistry informs diagnosis and clinical outcomes
- Author
-
Roxanna M. Garcia, Nitin R. Wadhwani, Rishi Lulla, Charles David James, Amanda Saratsis, Amir Behdad, Ali Shilatifard, Jin Qi, Craig Horbinski, and Tina Huang
- Subjects
K27m mutation ,business.industry ,histone H3K27M mutation ,diffuse intrinsic pontine glioma ,Correction ,medicine.disease ,histone H3 post-translational modification ,diffuse midline glioma ,Histone H3 ,Oncology ,Glioma ,Cancer research ,Posttranslational modification ,pediatric glioma ,Medicine ,Immunohistochemistry ,business ,Research Paper - Abstract
Pediatric diffuse midline glioma is a highly morbid glial neoplasm that may arise in the thalamus or brainstem (also known as diffuse intrinsic pontine glioma or DIPG). Because tumor anatomic location precludes surgical resection, diagnosis and treatment is based on MR imaging and analysis of biopsy specimens. Up to 80% of pediatric diffuse midline gliomas harbor a histone H3 mutation resulting in the replacement of lysine 27 with methionine (K27M) in genes encoding histone H3 variant H3.3 (H3F3A) or H3.1 (HIST1H3B). H3K27M mutant glioma responds more poorly to treatment and is associated with worse clinical outcome than wild-type tumors, so mutation detection is now diagnostic for a new clinical entity, diffuse midline glioma H3K27M mutant, as defined in the most recent WHO classification system. We previously reported patterns of histone H3 trimethylation (H3K27me3) and acetylation (H3K27Ac) associated with H3K27M mutation that impact transcription regulation and contribute to tumorigenesis. Given the clinical implications of the H3K27M mutation and these associated H3 post-translational modifications (PTMs), we set to determine whether they can be characterized via immunohistochemistry (IHC) in a cohort of pediatric glioma (n = 69) and normal brain tissue (n = 4) specimens. We observed 100% concordance between tissue IHC and molecular sequencing for detecting H3K27M mutation. In turn, H3K37M and H3K27me3 results, but not H3K27Ac staining patterns, were predictive of clinical outcomes. Our results demonstrate H3K27M and H3K27me3 staining of pediatric glioma tissue may be useful for diagnosis, stratification to epigenetic targeted therapies, and longitudinal monitoring of treatment response.
- Published
- 2019
46. HG-61MOLECULAR CHARACTERIZATION OF IN VIVO AND IN VITRO MODELS OF DIPG
- Author
-
Eric H. Raabe, Javad Nazarian, Amanda Saratsis, Mamta Giri, Sridevi Yadavilli, Roger J. Packer, Nalin Gupta, Kristy J. Brown, Madhuri Kambhampati, Kathy Warren, and Mojca Stampar
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Abstracts ,Tissue specimen ,Oncology ,In vivo ,Cancer research ,medicine ,Neurology (clinical) ,Biology ,In vitro - Published
- 2016
47. Mutations in chromatin machinery and pediatric high-grade glioma
- Author
-
Rishi Lulla, Rintaro Hashizume, and Amanda Saratsis
- Subjects
0301 basic medicine ,Transcriptional Activation ,Central nervous system ,JMJD3 ,Context (language use) ,Review ,demethylase ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Glioma ,medicine ,Gene silencing ,Animals ,Humans ,Epigenetics ,Gene Silencing ,Molecular Targeted Therapy ,G34V/R ,GSKJ4 ,Multidisciplinary ,biology ,Brain Neoplasms ,SciAdv r-articles ,Histone mutation ,medicine.disease ,PRC2 ,Chromatin ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Histone ,medicine.anatomical_structure ,K27M ,Cell Transformation, Neoplastic ,Oncology ,Mutation ,biology.protein ,Cancer research ,pediatric high-grade glioma ,DIPG ,methyltransferase ,Neoplasm Grading - Abstract
Mutations in chromatin machinery define pediatric high-grade gliomas; efforts to define and target their functions are under way., Pediatric central nervous system tumors are the most common solid tumor of childhood. Of these, approximately one-third are gliomas that exhibit diverse biological behaviors in the unique context of the developing nervous system. Although low-grade gliomas predominate and have favorable outcomes, up to 20% of pediatric gliomas are high-grade. These tumors are a major contributor to cancer-related morbidity and mortality in infants, children, and adolescents, with long-term survival rates of only 10 to 15%. The recent discovery of somatic oncogenic mutations affecting chromatin regulation in pediatric high-grade glioma has markedly improved our understanding of disease pathogenesis, and these findings have stimulated the development of novel therapeutic approaches targeting epigenetic regulators for disease treatment. We review the current perspective on pediatric high-grade glioma genetics and epigenetics, and discuss the emerging and experimental therapeutics targeting the unique molecular abnormalities present in these deadly childhood brain tumors.
- Published
- 2016
48. PDTM-02. NEXT-GENERATION SEQUENCING OF DIFFUSE INTRINSIC PONTINE GLIOMA CELLS REVEALS ALTERED EPIGENETIC REGULATION AND DISTINCT TUMOR SUBGROUPS
- Author
-
Rintaro Hashizume, Jin Qi, Elizabeth T. Bartom, Amanda Saratsis, Charles David James, Ali Shilatifard, Stacy A. Marshall, and Tina Huang
- Subjects
Cancer Research ,biology ,medicine.disease_cause ,Molecular biology ,DNA sequencing ,Chromatin ,Gene expression profiling ,Abstracts ,Histone ,Oncology ,Transcriptional regulation ,Cancer research ,biology.protein ,Neuron differentiation ,medicine ,Neurology (clinical) ,Epigenetics ,Carcinogenesis - Published
- 2017
49. PDTM-39. HISTONE H3 MUTATION EFFECTS ON CHROMATIN STRUCTURE AND REGULATION OF GENE TRANSCRIPTION IN PEDIATRIC GLIOMA
- Author
-
Elizabeth T. Bartom, Ali Shilatifard, Patrick A. Ozark, Stacy A. Marshall, Tina Huang, Amanda Saratsis, Andrea Piunti, C. David James, and Rintaro Hashizume
- Subjects
Cancer Research ,Mutation ,Biology ,medicine.disease_cause ,Chromatin ,Cell biology ,Abstracts ,Histone H3 ,Oncology ,Gene expression ,medicine ,Nucleosome ,Neurology (clinical) ,Epigenetics ,Transcription factor ,Gene - Abstract
INTRODUCTION: Pediatric high-grade glioma (HGG), including H3K27M diffuse midline glioma, has the highest mortality of pediatric solid tumors. Recurrent Histone H3 mutations result in methionine for lysine substitution (H3K27M) in 80% of diffuse midline gliomas, or valine for glycine/arginine substitution (G34V/R) in 50% of hemispheric HGGs. These mutations alter chromatin structure and are associated with distinct patterns of gene expression and poorer response to therapy. To elucidate the mechanism by which these mutations affect chromatin function, we characterized genomic deposition of Histone H3 proteoforms in a large cohort of rare pediatric glioma cell lines. METHODS: H3K27M DIPG (n=6), H3G34V (n=1) and wild-type pediatric high-grade glioma cells (n=2), neural stem cells (n=1) and astrocytes (n=1) were analyzed for genomic deposition patterns of H3.3, H3K27M, H3G34V, H3K27me3, H3K27Ac, H4Kme1, and H4Kme3. Extracted chromatins were sonicated to produce DNA fragments for ChIP. RNA was extracted for whole transcriptome profiling. DNA/RNA libraries were prepared using the KAPA HTP Library Preparation Kit and sequenced (ChIP-and RNA-Seq, Illumina NextSeq 500). Genomic enrichments and gene expressions were determined, quantified, and analyzed for biological relevance. RESULTS: Distinct genomic enrichment of H3 proteoforms was observed between mutant and wild type cell lines, corresponding with respective gene expression levels. Differential co-localization of H3 post-translational marks with mutant vs. wild type H3 protein was also observed, consistent with our prior description of mutant heterotypic nucleosomes. Location of specific marks (promoter, gene body) suggests altered transcription factor recruitment and function may result in observed patterns of gene expression. CONCLUSION: We present the largest known epigenetic analysis of pediatric glioma cell lines to date, indicating distinct patterns of Histone H3 enrichment and gene expression in H3K27M and H3G34V/R mutant lines. These data provide insight into the mechanisms by which H3 mutations impact pediatric glioma biology, which may inform novel therapeutic approaches.
- Published
- 2018
50. BT-01TENASCIN-C: A NOVEL THERAPEUTIC TARGET IN PEDIATRIC BRAINSTEM GLIOMA
- Author
-
Amanda Saratsis, Jordan Hall, Roger J. Packer, Guifa Xi, Javad Nazarian, Mojca Stampar, Madhuri Kambhampati, and Eric H. Raabe
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,Astrocytoma ,Biology ,musculoskeletal system ,medicine.disease ,Fold change ,Oncology ,Western blot ,Glioma ,DNA methylation ,Gene expression ,medicine ,Immunohistochemistry ,Neurology (clinical) ,Progenitor cell ,Abstracts from the 3rd Biennial Conference on Pediatric Neuro-Oncology Basic and Translational Research - Abstract
INTRODUCTION: Diffuse intrinsic pontine glioma (DIPG) is the most deadly solid tumor of childhood. Histone 3 mutation occurs in 80% of DIPGs, causing global transcriptional changes. Tenascin-C (TNC) is an extracellular matrix protein expressed during normal brain development by oligodendroglial progenitor cells (OPCs), the purported DIPG cell of origin. TNC is highly expressed in adult glioma, contributing to local invasion and poor survival. We report increased TNC in tumor tissue and cerebrospinal fluid (CSF) from children with high-grade glioma, including DIPG, and characterize TNC expression in relation to H3K27M mutation and DNA methylation status. METHODS: Tumor tissue collected intraoperatively or post-mortem from children with brainstem (DIPG, n = 14), supratentorial (n = 7), and cerebellar astrocytoma (n = 2), and CSF from DIPG (n = 9) and supratentorial astrocytoma (n = 17) was subjected to MS/MS proteomic analysis. Tissue gene expression, DNA methylation, H3F3A and HIST1H3B sequencing was performed. TNC expression was validated via Western blot and immunohistochemistry. Data integration was performed with Genome Studio, Partek and Ingenuity Pathway Analysis. RESULTS: TNC expression was significantly increased in 75% of gliomas compared to normal tissue, including all DIPG specimens tested (fold change >2, p < 0.05). Secreted TNC was detected in 7/9 DIPG CSF specimens (77.8%). Tumor-specific TNC expression was confirmed with Western blot and tissue immunohistochemistry. TNC expression correlated with tumor grade and was associated with Notch pathway activation, H3K27M mutation, and TNC promoter hypomethylation. CONCLUSIONS: We report increased TNC expression in tissue and CSF of pediatric high-grade gliomas, including DIPGs, associated with promoter hypomethylation and H3K27M mutation. Given the effect of TNC on OPC proliferation, migration and differentiation, TNC could serve as a clinical biomarker of disease and rational therapeutic target for a substantial subgroup of DIPG patients. Studies exploring the mechanism of TNC overexpression and effects of targeting TNC expression in DIPG are currently underway.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.